51
|
Zhang SQ, Thomas F, Fang J, Austgen K, Cowan C, Welstead GG. Universal protection of allogeneic T-cell therapies from natural killer cells via CD300a agonism. Blood Adv 2025; 9:254-264. [PMID: 39368806 PMCID: PMC11782830 DOI: 10.1182/bloodadvances.2024013436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 10/07/2024] Open
Abstract
ABSTRACT Immunogenicity limits the persistence of off-the-shelf allogeneic cell therapies and transplants. Although ablation of HLA removes most T cell and humoral alloreactivity, no solution has enabled universal protection against the resulting natural killer (NK) cell response. Here, we engineered trans-antigen signaling receptors (TASRs) as a new class of NK inhibitory ligands and discovered CD300a, a previously inaccessible receptor, as a functional target. CD300a TASR outperformed leading alternative strategies in focused screens, including CD47 and HLA-E, and was solely capable of universally protecting allogeneic T cells against a large human cohort (45/45 donors), spanning diverse demographics and NK cell phenotypes. A model allogeneic T-cell therapy coexpressing an anti-CD19 chimeric antigen receptor and CD300a TASR, produced using multiplexed nonviral integration, exhibited enhanced B-cell killing potency under allogeneic immune pressure. CD300 TASR represents a universal solution to NK alloreactivity, broadening the population that could be effectively treated by next-generation allogeneic cell therapies.
Collapse
Affiliation(s)
- Shu-Qi Zhang
- Department of Discovery Biology, Clade Therapeutics, Inc, Boston, MA
- Department of Molecular Immunology, Clade Therapeutics, Inc, Boston, MA
| | - Faith Thomas
- Department of Discovery Biology, Clade Therapeutics, Inc, Boston, MA
| | - Justin Fang
- Department of Discovery Biology, Clade Therapeutics, Inc, Boston, MA
- Department of Discovery Biology, Century Therapeutics, Inc, Boston, MA
- Department of Molecular Immunology, Clade Therapeutics, Inc, Boston, MA
| | - Kathryn Austgen
- Department of Molecular Immunology, Clade Therapeutics, Inc, Boston, MA
| | - Chad Cowan
- Clade Therapeutics, Inc, Boston, MA
- Century Therapeutics, Inc, Philadelphia, PA
| | - G. Grant Welstead
- Department of Discovery Biology, Clade Therapeutics, Inc, Boston, MA
- Department of Discovery Biology, Century Therapeutics, Inc, Boston, MA
| |
Collapse
|
52
|
Chiavarini M, Genga A, Ricciotti GM, D’Errico MM, Barbadoro P. Safety, Immunogenicity, and Efficacy of Cytomegalovirus Vaccines: A Systematic Review of Randomized Controlled Trials. Vaccines (Basel) 2025; 13:85. [PMID: 39852864 PMCID: PMC11768780 DOI: 10.3390/vaccines13010085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/10/2025] [Accepted: 01/15/2025] [Indexed: 01/26/2025] Open
Abstract
Background/Objectives: Cytomegalovirus (CMV) is widespread and mostly causes asymptomatic infections in immunocompetent hosts, but it may lead to severe and life-threatening diseases in immunocompromised individuals, such as transplant patients and congenitally infected children, representing a significant public health concern. Although there are no licensed CMV vaccines, the development of a CMV vaccine is considered a high priority due to its potential to reduce the burden associated with CMV-related complications, and several approaches are under investigation. The objective of this systematic review was to synthesize the evidence on various CMV vaccines currently under clinical development. Methods: According to the PRISMA guidelines (PROSPERO ID: CRD42024516601), a comprehensive literature search was conducted to identify all the randomized controlled trials that have evaluated the safety, immunogenicity, and efficacy of vaccine candidates compared to a placebo. A total of 26 studies were identified: 11 on transplant patients and 15 on healthy individuals. Results: Several vaccine candidates have shown encouraging results in terms of safety and specific immune responses, notably adjuvanted gB vaccines and DNA vaccines targeting gB and pp65. The results were divided into RCTs on healthy individuals and those on transplant recipients, because the CMV-specific immune response to a vaccine is complex and varies depending not only on the type of vaccine, but also on the immunological status of the individual. Conclusions: Challenges remain in achieving broad efficacy across diverse populations, particularly for immunocompromised patients. Thus, the present work seeks to support future decisions and guide further research in the development of an effective and widely available CMV vaccine.
Collapse
Affiliation(s)
- Manuela Chiavarini
- Department of Health Sciences, University of Florence, Viale GB Morgagni 48, 50134 Florence, Italy;
| | - Anita Genga
- Department of Biomedical Sciences and Public Health, Section of Hygiene, Preventive Medicine and Public Health, Polytechnic University of the Marche Region, 60126 Ancona, Italy; (M.M.D.); (P.B.)
| | - Giorgia Maria Ricciotti
- Department of Biomedical Sciences and Public Health, Section of Hygiene, Preventive Medicine and Public Health, Polytechnic University of the Marche Region, 60126 Ancona, Italy; (M.M.D.); (P.B.)
| | - Marcello Mario D’Errico
- Department of Biomedical Sciences and Public Health, Section of Hygiene, Preventive Medicine and Public Health, Polytechnic University of the Marche Region, 60126 Ancona, Italy; (M.M.D.); (P.B.)
| | - Pamela Barbadoro
- Department of Biomedical Sciences and Public Health, Section of Hygiene, Preventive Medicine and Public Health, Polytechnic University of the Marche Region, 60126 Ancona, Italy; (M.M.D.); (P.B.)
| |
Collapse
|
53
|
Neugebauer E, Walter S, Tan J, Drayman N, Franke V, van Gent M, Pennisi S, Veratti P, Stein KS, Welker I, Tay S, Verjans GMGM, Timmers HTM, Akalin A, Landthaler M, Ensser A, Wyler E, Full F. Herpesviruses mimic zygotic genome activation to promote viral replication. Nat Commun 2025; 16:710. [PMID: 39814710 PMCID: PMC11735616 DOI: 10.1038/s41467-025-55928-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 01/05/2025] [Indexed: 01/18/2025] Open
Abstract
Zygotic genome activation (ZGA) is crucial for maternal to zygotic transition at the 2-8-cell stage in order to overcome silencing of genes and enable transcription from the zygotic genome. In humans, ZGA is induced by DUX4, a pioneer factor that drives expression of downstream germline-specific genes and retroelements. Here we show that herpesviruses from all subfamilies, papillomaviruses and Merkel cell polyomavirus actively induce DUX4 expression to promote viral transcription and replication. Analysis of single-cell sequencing data sets from patients shows that viral DUX4 activation is of relevance in vivo. Herpes-simplex virus 1 (HSV-1) immediate early proteins directly induce expression of DUX4 and its target genes, which mimics zygotic genome activation. Upon HSV-1 infection, DUX4 directly binds to the viral genome and promotes viral transcription. DUX4 is functionally required for infection, since genetic depletion by CRISPR/Cas9 as well as degradation of DUX4 by nanobody constructs abrogates HSV-1 replication. Our results show that DNA viruses including herpesviruses mimic an embryonic-like transcriptional program that prevents epigenetic silencing of the viral genome and facilitates herpesviral gene expression.
Collapse
Affiliation(s)
- Eva Neugebauer
- Institute of Virology, University Medical Center, and Faculty of Medicine, Albert-Ludwig-University Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Stephanie Walter
- Institute for Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Jiang Tan
- Institute of Virology, University Medical Center, and Faculty of Medicine, Albert-Ludwig-University Freiburg, Freiburg, Germany
| | - Nir Drayman
- The Department of Molecular Biology and Biochemistry, the Center for Virus Research and the Center for Complex Biological Systems, The University of California, Irvine, Irvine, CA, 92697, USA
| | - Vedran Franke
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Helmholtz Society, Berlin, Germany
| | - Michiel van Gent
- HerpesLabNL, Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Sandra Pennisi
- Institute of Virology, University Medical Center, and Faculty of Medicine, Albert-Ludwig-University Freiburg, Freiburg, Germany
| | - Pia Veratti
- Institute of Virology, University Medical Center, and Faculty of Medicine, Albert-Ludwig-University Freiburg, Freiburg, Germany
| | - Karla S Stein
- Institute of Virology, University Medical Center, and Faculty of Medicine, Albert-Ludwig-University Freiburg, Freiburg, Germany
| | - Isabelle Welker
- Institute of Virology, University Medical Center, and Faculty of Medicine, Albert-Ludwig-University Freiburg, Freiburg, Germany
| | - Savaş Tay
- The Pritzker School for Molecular Engineering, The University of Chicago, Chicago, IL, 60637, USA
| | - Georges M G M Verjans
- HerpesLabNL, Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - H T Marc Timmers
- German Cancer Consortium (DKTK), partner site Freiburg, a partnership between the DKFZ and Medical Center-University of Freiburg, and Department of Urology, Medical Center-University of Freiburg, Freiburg, Germany
| | - Altuna Akalin
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Helmholtz Society, Berlin, Germany
| | - Markus Landthaler
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Helmholtz Society, Berlin, Germany
| | - Armin Ensser
- Institute for Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Emanuel Wyler
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Helmholtz Society, Berlin, Germany
| | - Florian Full
- Institute of Virology, University Medical Center, and Faculty of Medicine, Albert-Ludwig-University Freiburg, Freiburg, Germany.
- German Consulting Laboratory for HSV and VZV, Medical Center - University of Freiburg, Freiburg, Germany.
| |
Collapse
|
54
|
Vernooij RW, Michael M, Colombijn JM, Owers DS, Webster AC, Strippoli GF, Hodson EM. Pre-emptive treatment for cytomegalovirus viraemia to prevent cytomegalovirus disease in solid organ transplant recipients. Cochrane Database Syst Rev 2025; 1:CD005133. [PMID: 39807668 PMCID: PMC11729901 DOI: 10.1002/14651858.cd005133.pub4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
BACKGROUND Cytomegalovirus (CMV) is a significant cause of morbidity and death in solid organ transplant recipients. Pre-emptive treatment of patients with CMV viraemia using antiviral agents has been suggested as an alternative to routine prophylaxis to prevent CMV disease. This is an update of a Cochrane review first published in 2006 and updated in 2013. OBJECTIVES To determine the benefits and harms of pre-emptive treatment of CMV viraemia to prevent CMV disease and death (any cause) and the indirect effects of CMV infection (acute rejection, graft loss, opportunistic infections) in solid organ transplant recipients. SEARCH METHODS The Cochrane Kidney and Transplant Register of Studies was searched up to 17 December 2024 using search terms relevant to this review. Studies in the Register are identified through searches of CENTRAL, MEDLINE, and EMBASE, conference proceedings, the International Clinical Trials Registry Platform (ICTRP) Search Portal, and ClinicalTrials.gov. SELECTION CRITERIA We included randomised controlled trials (RCTs) and quasi-RCTs comparing pre-emptive treatment with placebo, no specific treatment, or antiviral prophylaxis in solid organ transplant recipients. DATA COLLECTION AND ANALYSIS Two authors independently assessed the eligibility of the identified studies, assessed the risk of bias, and extracted all data. Results were expressed as risk ratio (RR) and 95% confidence intervals (CI) for dichotomous outcomes. Statistical analyses were performed using a random-effects model. The certainty of evidence was assessed per outcome using the Grades of Recommendation, Assessment, Development and Evaluation (GRADE) approach. MAIN RESULTS In this update, we have included seven new studies, bringing the total number of included studies to 22 (1883 participants). Of these, seven investigated pre-emptive treatment versus placebo or standard care, 12 looked at pre-emptive treatment versus antiviral prophylaxis, one study investigated oral versus intravenous pre-emptive treatment, one investigated pre-emptive valganciclovir versus pre-emptive ganciclovir, and one investigated letermovir 40 mg twice/day versus 80 mg once/day. Studies were conducted in Australia, Brazil, the Czech Republic, Germany, Italy, Japan, Norway, Spain, South Korea, and the USA. Organ transplant recipients included kidney, liver, heart, lung, and kidney-pancreas. Thirteen studies were single-centre studies, six were multicentre, and three were unknown. The number of participants ranged from 12 to 296. Overall, selection bias was unclear (55%); performance, detection and attrition bias were high (91%, 63% and 95%, respectively), and reporting bias was low (55%). Compared with placebo or standard care, pre-emptive treatment probably reduces the risk of CMV disease (7 studies, 315 participants: RR 0.29, 95% CI 0.11 to 0.80; I2 = 54%; moderate-certainty evidence) but may result in little or no difference in death (any cause) (3 studies, 176 participants: RR 1.23, 95% CI 0.35 to 4.30; I2 = 0%; low-certainty evidence). Pre-emptive treatment may result in little or no difference in CMV organ involvement, CMV-associated symptoms, acute rejection, graft loss, other infections or leucopenia. Compared to prophylaxis, pre-emptive treatment may make little or no difference to the risk of developing CMV disease (11 studies, 1322 participants: RR 0.97, 95% CI 0.47 to 2.01; I2 = 54%; low-certainty evidence) and probably makes little or no difference to death (any cause) (9 studies, 1098 participants: RR 0.95, 95% CI 0.60 to 1.52; I2 = 0%; moderate-certainty evidence). Pre-emptive treatment may increase the risk of CMV infection (8 studies, 867 participants: RR 1.97, 95% CI 1.48 to 2.61; I2 = 66%; low-certainty evidence). The risk of leucopenia (7 studies, 869 participants: RR 0.57, 95% CI 0.38 to 0.87; I2 = 33%; moderate-certainty evidence) and neutropenia (5 studies, 859 participants: RR 0.63, 95% CI 0.44 to 0.90; I2 = 0% moderate certainty evidence) probably decreases with pre-emptive therapy. There may be little or no difference in the risks of acute rejection, graft loss, and infections other than CMV. Single studies were identified for comparisons between different pre-emptive treatments: 1) oral ganciclovir versus IV ganciclovir; 2) valganciclovir versus ganciclovir; 3) 40 mg twice/day versus 80 mg once/day. No differences between these treatment modalities in terms of CMV disease, death (any cause), or adverse events were identified. AUTHORS' CONCLUSIONS In this review, we have included seven new studies, yet the available evidence is overall of low certainty and the conclusions remain similar to the previous version of this review. Pre-emptive treatment probably reduces the risk of CMV disease compared with placebo or standard care. There were no clear differences between pre-emptive treatment and prophylaxis to prevent CMV disease or reduce the risk of death (any cause). The risk of CMV infection may be higher for patients receiving pre-emptive therapy, but the risk of adverse events, such as leucopenia, is probably lower.
Collapse
Affiliation(s)
- Robin Wm Vernooij
- Department of Nephrology and Hypertension and Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, Netherlands
| | - Mini Michael
- Division of Pediatric Nephrology, Department of Pediatrics, Texas Children's Hospital/Baylor College of Medicine, Houston, TX, USA
| | - Julia Mt Colombijn
- Department of Nephrology and Hypertension and Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, Netherlands
| | - Daniel S Owers
- Department of Critical Care, The Canberra Hospital, Garran, Australia
| | - Angela C Webster
- Sydney School of Public Health, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Westmead Applied Research Centre, The University of Sydney at Westmead, Westmead, Australia
- Department of Transplant and Renal Medicine, Westmead Hospital, Westmead, Australia
| | - Giovanni Fm Strippoli
- Department of Precision and Regenerative Medicine and Ionian Area (Dimepre-J), University of Bari, Bari, Italy
- Sydney School of Public Health, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Cochrane Kidney and Transplant, Centre for Kidney Research, The Children's Hospital at Westmead, Westmead, Australia
| | - Elisabeth M Hodson
- Cochrane Kidney and Transplant, Centre for Kidney Research, The Children's Hospital at Westmead, Westmead, Australia
| |
Collapse
|
55
|
Bruton J, Hanke T. Exploitation of Unconventional CD8 T-Cell Responses Induced by Engineered Cytomegaloviruses for the Development of an HIV-1 Vaccine. Vaccines (Basel) 2025; 13:72. [PMID: 39852851 PMCID: PMC11769474 DOI: 10.3390/vaccines13010072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/08/2025] [Accepted: 01/12/2025] [Indexed: 01/26/2025] Open
Abstract
After four decades of intensive research, traditional vaccination strategies for HIV-1 remain ineffective due to HIV-1's extraordinary genetic diversity and complex immune evasion mechanisms. Cytomegaloviruses (CMV) have emerged as a novel type of vaccine vector with unique advantages due to CMV persistence and immunogenicity. Rhesus macaques vaccinated with molecular clone 68-1 of RhCMV (RhCMV68-1) engineered to express simian immunodeficiency virus (SIV) immunogens elicited an unconventional major histocompatibility complex class Ib allele E (MHC-E)-restricted CD8+ T-cell response, which consistently protected over half of the animals against a highly pathogenic SIV challenge. The RhCMV68-1.SIV-induced responses mediated a post-infection replication arrest of the challenge virus and eventually cleared it from the body. These observations in rhesus macaques opened a possibility that MHC-E-restricted CD8+ T-cells could achieve similar control of HIV-1 in humans. The potentially game-changing advantage of the human CMV (HCMV)-based vaccines is that they would induce protective CD8+ T-cells persisting at the sites of entry that would be insensitive to HIV-1 evasion. In the RhCMV68-1-protected rhesus macaques, MHC-E molecules and their peptide cargo utilise complex regulatory mechanisms and unique transport patterns, and researchers study these to guide human vaccine development. However, CMVs are highly species-adapted viruses and it is yet to be shown whether the success of RhCMV68-1 can be translated into an HCMV ortholog for humans. Despite some safety concerns regarding using HCMV as a vaccine vector in humans, there is a vision of immune programming of HCMV to induce pathogen-tailored CD8+ T-cells effective against HIV-1 and other life-threatening diseases.
Collapse
Affiliation(s)
- Joseph Bruton
- Hertford College, University of Oxford, Oxford OX1 3BW, UK;
| | - Tomáš Hanke
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK
| |
Collapse
|
56
|
Venturini C, Breuer J. Cytomegalovirus Genetic Diversity and Evolution: Insights into Genotypes and Their Role in Viral Pathogenesis. Pathogens 2025; 14:50. [PMID: 39861011 PMCID: PMC11768282 DOI: 10.3390/pathogens14010050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 12/20/2024] [Accepted: 12/24/2024] [Indexed: 01/27/2025] Open
Abstract
Cytomegalovirus (CMV) is a ubiquitous virus that infects most of the human population and causes significant morbidity and mortality, particularly among immunocompromised individuals. Understanding CMV's genetic diversity and evolutionary dynamics is crucial for elucidating its pathogenesis and developing effective therapeutic interventions. This review provides a comprehensive examination of CMV's genetic diversity and evolution, focussing on the role of different genotypes in viral pathogenesis.
Collapse
Affiliation(s)
- Cristina Venturini
- Department of Infection, Immunity and Inflammation, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK;
| | | |
Collapse
|
57
|
Permar SR, Schleiss MR, Plotkin SA. A vaccine against cytomegalovirus: how close are we? J Clin Invest 2025; 135:e182317. [PMID: 39744948 DOI: 10.1172/jci182317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
The pursuit of a vaccine against the human cytomegalovirus (HCMV) has been ongoing for more than 50 years. HCMV is the leading infectious cause of birth defects, including damage to the brain, and is a common cause of complications in organ transplantation. The complex biology of HCMV has made vaccine development difficult, but a recent meeting sponsored by the National Institute of Allergy and Infectious Diseases in September of 2023 brought together experts from academia, industry, and federal agencies to discuss progress in the field. The meeting reviewed the status of candidate HCMV vaccines under study and the challenges in clinical trial design in demonstrating efficacy against congenital CMV infection or the reduction of HCMV disease following solid organ transplantation or hematopoietic stem cell transplantation. Discussion in the meeting revealed that, with the numerous candidate vaccines that are under study, it is clear that a safe and effective HCMV vaccine is within reach. Meeting attendees achieved a consensus opinion that even a partially effective vaccine would have a major effect on the global health consequences of HCMV infection.
Collapse
Affiliation(s)
- Sallie R Permar
- Department of Pediatrics, Weill Cornell Medical Center, New York, New York, USA
| | - Mark R Schleiss
- Division of Pediatric Infectious Diseases and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Stanley A Plotkin
- Department of Pediatrics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Vaxconsult, Doylestown, Pennsylvania, USA
| |
Collapse
|
58
|
Chun Heng L, Muhd Nor NI. Management of Glaucoma in Cytomegalovirus Uveitis: A Retrospective Case Series of Surgical and Medical Approaches. Cureus 2025; 17:e76946. [PMID: 39906435 PMCID: PMC11793078 DOI: 10.7759/cureus.76946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/05/2025] [Indexed: 02/06/2025] Open
Abstract
Cytomegalovirus (CMV) is a double-stranded (dsDNA) virus of the herpesvirus family. Serological tests reveal signs of previous exposure to it in 40% to 100% of the general population. CMV anterior uveitis (AU) is the most common form of ocular manifestation of CMV in immunocompetent individuals. Clinically, it manifests mainly as anterior chamber (AC) inflammation, iris atrophy, and elevated intraocular pressure (IOP). In this study, we presented four cases of CMV AU with high IOP requiring different treatment modalities to control the IOP. All patients underwent AC paracentesis, and the aqueous sample sent for polymerase chain reaction (PCR) showed CMV DNA. They were treated with ganciclovir ophthalmic gel 0.15% for the infection. For IOP control, patients underwent different surgeries, namely microinvasive glaucoma surgery (MIGS) with XEN implant, augmented trabeculectomy (AT), glaucoma drainage device implantation (GDI), and transscleral cyclophotocoagulation (TSCPC). Patients were then followed up for a period ranging from six months to three years post-intervention to monitor for evidence of recurrence, IOP control, number of topical antiglaucoma medications required, and progression of glaucoma as evidenced by optical coherence tomography (OCT) retina nerve fiber layer (RNFL) and Humphrey visual field (HVF).
Collapse
Affiliation(s)
- Low Chun Heng
- Ophthalmology, Hospital Kuala Lumpur, Kuala Lumpur, MYS
| | | |
Collapse
|
59
|
Atanasoff KE, Ophir SI, Parsons AJ, Paredes Casado J, Lurain NS, Bowlin TL, Opperman TJ, Tortorella D. N-arylpyrimidinamine (NAPA) compounds are broadly acting inhibitors of human cytomegalovirus infection and spread. Antiviral Res 2025; 233:106044. [PMID: 39608645 DOI: 10.1016/j.antiviral.2024.106044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 11/20/2024] [Accepted: 11/25/2024] [Indexed: 11/30/2024]
Abstract
Human cytomegalovirus (HCMV) is a β-herpesvirus that contributes to the disease burden of immunocompromised and immunomodulated individuals, including transplant recipients and newborns. The FDA-approved HCMV drugs can exhibit drug resistance and severe side effects including bone marrow toxicity, gastrointestinal disruption, and nephrotoxicity. In a previous study, we identified the N-arylpyrimidinamine (NAPA) compound series as a new class of HCMV inhibitors that target early stages of infection. Here we describe the inhibitory activity of two potent NAPA analogs, MBXC-4336 and MBX-4992, that broadly block infection and spread. MBXC-4336 and MBX-4992 effectively inhibited infection by diverse HCMV strains and significantly prevented virus spread in fibroblast and epithelial cells as evaluated by quantifying infected cells and viral genome levels. Further, the NAPA compounds limited replication of clinical HCMV isolates, including a ganciclovir-resistant strain. Importantly, combination studies of NAPA compounds with ganciclovir demonstrated additive or synergistic inhibition of HCMV spread. Collectively, NAPA compounds have therapeutic potential for development as a novel class of anti-HCMV drugs.
Collapse
Affiliation(s)
- Kristina E Atanasoff
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Sabrina I Ophir
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Andrea J Parsons
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Jailene Paredes Casado
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Nell S Lurain
- Department of Immunology-Microbiology, Rush University, Chicago, IL, USA
| | | | | | - Domenico Tortorella
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
60
|
Maessen L, Boers LS, Heylen J, van Someren Gréve F, Wauters J, Bos LDJ, Feys S. Viral reactivations and fungal infections in nonresolving acute respiratory distress syndrome. Eur Respir Rev 2025; 34:240153. [PMID: 39971398 PMCID: PMC11836671 DOI: 10.1183/16000617.0153-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 11/21/2024] [Indexed: 02/21/2025] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a condition affecting 10% of patients requiring admission to the intensive care unit and results from endothelial dysfunction, alveolar epithelial injury and unbalanced inflammation, leading to exudative pulmonary oedema. A significant portion of these patients experience a lung injury that fails to resolve. Persistent or worsening respiratory failure beyond 5 days after the initiation of mechanical ventilation is referred to as nonresolving ARDS. Viral and fungal pathogens can exploit the hyperinflammatory environment and altered immune landscape in ARDS, perpetuating a cycle of ongoing inflammation and lung injury, thereby contributing to the progression towards and persistence of nonresolving ARDS, even in previously immunocompetent patients. This review discusses the significance, pathophysiology, diagnostic challenges and key knowledge gaps concerning various viral and fungal pathogens in nonresolving ARDS, with a particular focus on influenza-associated and COVID-19-associated pulmonary aspergillosis and pulmonary reactivation of Herpesviridae, such as cytomegalovirus and herpes simplex virus. Diagnosing these infections is challenging due to their nonspecific clinical presentation and the inability of current tests to distinguish between fungal colonisation or asymptomatic viral shedding and clinically significant infections or reactivations. A deeper understanding of the complex interplay between these pathogens and the host immune system in the context of ARDS, combined with advances in diagnostic and therapeutic strategies, has the potential to enhance the management and prognosis of patients with nonresolving ARDS.
Collapse
Affiliation(s)
- Lenn Maessen
- Medical Intensive Care Unit, Department of Internal Medicine, University Hospitals Leuven, Leuven, Belgium
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
- Shared first authors
| | - Leonoor S Boers
- Department of Intensive Care Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Shared first authors
| | - Jannes Heylen
- Medical Intensive Care Unit, Department of Internal Medicine, University Hospitals Leuven, Leuven, Belgium
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Frank van Someren Gréve
- Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Joost Wauters
- Medical Intensive Care Unit, Department of Internal Medicine, University Hospitals Leuven, Leuven, Belgium
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
- Shared last authors
| | - Lieuwe D J Bos
- Department of Intensive Care Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Shared last authors
| | - Simon Feys
- Medical Intensive Care Unit, Department of Internal Medicine, University Hospitals Leuven, Leuven, Belgium
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
- Shared last authors
| |
Collapse
|
61
|
Borbye-Lorenzen N, Holmgaard S, Ottosson F, Nudel R, Appadurai V, Laursen TM, Bækvad-Hansen M, Bybjerg-Grauholm J, Nordentoft M, Børglum AD, Mortensen PB, Werge T, Benros ME, Hougaard DM, Skogstrand K. High level of immunoglobulin G targeting mycoplasma or cytomegalovirus in the newborn increases risk of ADHD. Brain Behav Immun 2025; 123:99-107. [PMID: 39260764 DOI: 10.1016/j.bbi.2024.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 08/26/2024] [Accepted: 09/07/2024] [Indexed: 09/13/2024] Open
Abstract
Attention-deficit hyperactivity disorder (ADHD) is a neurodevelopmental disorder typically detected in childhood. Although ADHD has been demonstrated to have a strong genetic component, environmental risk factors, such as maternal infections during pregnancy, may also play a role. We therefore measured the immunological response to 5 abundant microorganisms (Toxoplasmosis Gondii, cytomegalovirus (CMV), Herpes Simplex Virus 1, Epstein Barr Virus and mycoplasma pneumoniae) in newborn heel prick samples of 1679 ADHD cases and 2948 matching controls as part of the iPSYCH Danish case-cohort study. We found an association between high anti-CMV (OR 1.30, 95 % CI [1.09,1.55], p = 0.015) and anti-mycoplasma (OR 1.30, 95 % CI [1.07,1.59], p = 0.037) signal and those newborns later being diagnosed with ADHD. The risk estimate remained increased when controlling for ADHD polygenic risk score as well as penicillin prescriptions. We saw a dose-response association with the amount of positive anti-microorganism titers increasing the risk of being diagnosed with ADHD later in life (p = 0.01 for the trend), suggesting that the more activated the immune system is prior to or at birth, the higher the risk is for a later diagnosis with ADHD. If the associations are causal, they emphasize the importance of a healthy life style during pregnancy to reduce the risk of infections when pregnant and the associated risks for the child.
Collapse
Affiliation(s)
- Nis Borbye-Lorenzen
- Danish Center for Neonatal Screening, Department of Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Solveig Holmgaard
- Danish Center for Neonatal Screening, Department of Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Filip Ottosson
- Danish Center for Neonatal Screening, Department of Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Ron Nudel
- Copenhagen Research Center for Biological and Precision Psychiatry, Mental Health Centre Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark; CORE - Copenhagen Research Center for Mental Health, Mental Health Centre Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark; iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
| | - Vivek Appadurai
- Institute of Biological Psychiatry, Mental Health Services, Copenhagen University Hospital, Copenhagen, Denmark
| | - Thomas Munk Laursen
- NCRR, National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark
| | - Marie Bækvad-Hansen
- Danish Center for Neonatal Screening, Department of Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Jonas Bybjerg-Grauholm
- Danish Center for Neonatal Screening, Department of Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Merete Nordentoft
- CORE - Copenhagen Research Center for Mental Health, Mental Health Centre Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark; iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anders D Børglum
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark; Department of Biomedicine - Human Genetics, Aarhus University, Aarhus, Denmark; Center for Genomics and Personalized Medicine, Aarhus, Denmark
| | - Preben Bo Mortensen
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark; NCRR, National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark
| | - Thomas Werge
- Institute of Biological Psychiatry, Mental Health Services, Copenhagen University Hospital, Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael E Benros
- Copenhagen Research Center for Biological and Precision Psychiatry, Mental Health Centre Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - David Michael Hougaard
- Danish Center for Neonatal Screening, Department of Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Kristin Skogstrand
- Danish Center for Neonatal Screening, Department of Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark.
| |
Collapse
|
62
|
Xie J, Shang L, Liu C, Mao J, He C, Luo M, Fisher D, Thi Thu Hien N, Xu S, Zhao L. Corilagin inhibits human cytomegalovirus infection and replication via activating the cGAS-STING signaling pathway in vitro and in vivo. Int Immunopharmacol 2024; 143:113401. [PMID: 39423664 DOI: 10.1016/j.intimp.2024.113401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 10/21/2024]
Abstract
AIM The existence of human cytomegalovirus (HCMV) is extremely widespread, causing serious diseases in patients with low immune function. The purpose of this study is to explore the efficacy and mechanism of Corilagin in the control of CMV infection, in order to provide scientific basis for the control of CMV infection. METHODS Our study employed an animal model in Balb/c mice, infected with MCMV, alongside cellular models in HFF cells and THP-1 cells, stimulated with HCMV. The expression of cGAS-STING signaling pathway molecules was detected in liver tissue, lung tissue, serum, cells and cell supernatant. The liver function and histopathological changes of mice were evaluated. RESULTS In vivo and in vitro experiments showed that Corilagin significantly inhibits CMV levels and attenuates pathological damage in liver and lung tissues in vivo, and similarly inhibits viral load in cells in vitro. Corilagin promotes the expression levels of STING and its downstream molecules in vivo and in vitro. Inhibition/down-regulation of STING significantly promotes CMV replication, on the contrary, activation/up-regulation of STING inhibits CMV replication, and Corilagin also promotes the expression levels of molecules related to the cGAS-STING signaling pathway in the above cases. CONCLUSION Corilagin could effectively inhibit the infection and replication of CMV in vitro and in vivo, which may be through the activation of cGAS-STING signaling pathway.
Collapse
Affiliation(s)
- Jiao Xie
- Health Management Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Luorui Shang
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cuiling Liu
- The Tenth Affiliated Hospital of Southern Medical University, Dongguan, China
| | - Jinqian Mao
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chengyi He
- Department of Vascular Surgery, the Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Miao Luo
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - David Fisher
- Department of Medical Biosciences, Faculty of Natural Sciences, University of The Western Cape, Cape Town, South Africa
| | | | - Sanping Xu
- Health Management Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Lei Zhao
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
63
|
Zhang Z, Zhang J, Dai S, Fan X, Liu Y, Sun J, Chen L, Song T, Li S, Zhang J, Liu X, Zhang R, Liu D, Xu Y, Li Y, Liu X. Active cytomegalovirus infection in mechanically ventilated patients with sepsis. BMC Infect Dis 2024; 24:1405. [PMID: 39696007 PMCID: PMC11654325 DOI: 10.1186/s12879-024-10304-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Active cytomegalovirus (CMV) infection is associated with poor prognosis in septic patients with critical illness. Patients of septic category are highly likely to benefit from prophylactic antiviral therapy. Nevertheless, the clinical characteristics for CMV reactivation are lacking among septic patients requiring mechanical ventilation. The aim of this study was to investigate the incidence, risk factors, and clinical outcomes regarding active CMV infection in mechanically ventilated patients with sepsis. METHODS A single-center, retrospective cohort study conducted from January 2021 to December 2023 that included septic patients on mechanical ventilation at the intensive care unit (ICU) of a national hospital. Study participants were divided into active and non-active CMV infection groups based on CMV DNAemia within a 28-day hospitalization period in ICU. Clinical features, laboratory findings, treatment measures, and clinical outcomes were compared between the two groups. RESULTS Among 118 septic patients, 21 (17.8%) exhibited active CMV infection within 28-day ICU admission. Hemoglobin served as an independent risk factor and predictor for active CMV infection (P < 0.05). Moreover, the duration of mechanical ventilation and ICU stay in active CMV infection patients were significantly higher than in the comparison group (P < 0.05). CONCLUSIONS Active CMV infection is common and associated with adverse clinical outcomes in mechanically ventilated patients with sepsis. A low level of hemoglobin is an independent risk factor for active CMV infection. Further prospective studies are warranted to assess the efficacy of initiating prophylactic and preemptive antiviral therapies among patients with sepsis disorders.
Collapse
Affiliation(s)
- Zhihui Zhang
- Department of Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Jierong Zhang
- Department of Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Shuang Dai
- Department of Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Xueying Fan
- Department of Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Yuhua Liu
- Department of Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Junlu Sun
- Department of Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Lisheng Chen
- Department of Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Tiantian Song
- Department of Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Shangzuo Li
- Department of Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Jinjiang Zhang
- Department of Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Xuesong Liu
- Department of Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Rong Zhang
- Department of Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Dongdong Liu
- Department of Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Yonghao Xu
- Department of Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Yimin Li
- Department of Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China.
| | - Xiaoqing Liu
- Department of Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China.
| |
Collapse
|
64
|
Kalaria A, Mehta RB, Sood P, Zhang X, Morford HJ, Potluri V, Bridges JFP, Puttarajappa CM. Mitigating High-risk EBV and CMV Through Kidney Paired Donation: A Survey of Potential Donor and Recipient Candidates. Transplant Direct 2024; 10:e1737. [PMID: 39563724 PMCID: PMC11576024 DOI: 10.1097/txd.0000000000001737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/27/2024] [Accepted: 09/30/2024] [Indexed: 11/21/2024] Open
Abstract
Background High-risk cytomegalovirus (CMV) and Epstein-Barr virus (EBV) mismatches (ie, seropositive donors to seronegative recipients) among kidney transplant recipients lead to increased healthcare utilization, inferior allograft outcomes, and high mortality. We assessed the interest among prospective kidney donor and recipient candidates to participate in kidney paired donation (KPD) for averting CMV/EBV high-risk mismatches. Methods We surveyed 51 potential living donors and 102 kidney recipient candidates presenting for their evaluation visit at the University of Pittsburgh Medical Center between October 2022 and May 2023. We evaluated their general inclination toward KPD and their interest in KPD under various risk-benefit scenarios, particularly emphasizing the mitigation of high-risk mismatches associated with EBV and CMV. This was done using a 5-point Likert scale (1-low interest; 5-high interest) customized survey. Results There was high interest in KPD among both donor and recipient candidates (median score 4 versus 4; P = 0.09). However, donor candidates had a lower interest in KPD if they were compatible with their intended recipient (median score 2 versus 4; P < 0.001). Most donor (80.4%; N = 41) and recipient candidates (89.2%; N = 91) expressed a strong willingness to participate in KPD to prevent high-risk CMV and EBV mismatches, but this interest declined with longer transplant delays. Interest also varied on the basis of participants' income and employment status. Conclusions Interest in KPD for avoiding CMV and EBV was high among both donor and recipient candidates. Additional research is required to assess the capacity and desirability for KPD expansion, particularly among ABO and HLA-compatible pairs.
Collapse
Affiliation(s)
- Arjun Kalaria
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Rajil B Mehta
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Puneet Sood
- Division of Nephrology, Department of Medicine, University of California, San Francisco, CA
| | - Xingyu Zhang
- School of Health and Rehabilitation Sciences, University of Pittsburgh, Pittsburgh, PA
| | - Harry J Morford
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Vishnu Potluri
- Renal-Electrolyte and Hypertension Division, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - John F P Bridges
- Department of Biomedical Informatics, The Ohio State University College of Medicine, Columbus, OH
| | - Chethan M Puttarajappa
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, PA
| |
Collapse
|
65
|
Dhital R, Flint K, Kaptsan I, Hegde S, Daloul R, Shimamura M. Virus-specific Th17 Cells Are Induced by Human Cytomegalovirus after Renal Transplantation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1703-1712. [PMID: 39423238 PMCID: PMC11573647 DOI: 10.4049/jimmunol.2300742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 09/17/2024] [Indexed: 10/21/2024]
Abstract
CMV infection and Th17 cells are independently associated with increased risk for late allograft loss after renal transplantation. Although CMV-specific Th17 cells are detectable in animal models and nontransplant clinical populations, evidence linking CMV and Th17 cells after renal transplantation remains unclear. This prospective observational study evaluated a cohort of renal transplant recipients during 12 mo posttransplant to assess the presence of CMV-specific Th17 cells in peripheral blood and their relationship to pretransplant CMV serostatus and CMV DNAemia. CMV-specific Th17 cells were identified among CMV serostatus donor (D)+ and/or recipient (R)+ recipients and expanded during both primary (D+/R-) and reactivated (D+/R+, D-/R+) CMV DNAemia. A subset of CMV-specific Th17 cells coexpressed IFN-γ, indicating a Th1/17 phenotype. These Th17 and Th1/17 cells expressed CCR6, CCR5, activation and terminal differentiation markers (CD95, OX40, HLA-DR, CD57), and a central/effector memory phenotype. CMV-specific Th1/17 cells expressed activating/inhibitory receptors (CD57, 4-1BB, CD160, CTLA-4, PD-1) at higher frequencies than Th17 cells. In contrast, staphylococcal enterotoxin B-induced Th17 cells did not expand during CMV DNAemia, did not differ between CMV serostatus groups over time, expressed CCR6, predominantly coexpressed TNF-α, and had lower expression of activating and inhibitory receptors than pp65-specific Th17 and Th1/17 cells. These data show that CMV-specific Th17 cells expand during episodes of CMV DNAemia among renal transplant recipients, and that these virus-specific Th17 and Th1/17 cells have distinct phenotypes from global circulating Th(1)/17 cells. These results suggest a potential proinflammatory pathway by which CMV-induced Th17 cells may contribute to allograft injury, increasing risk for late allograft loss.
Collapse
Affiliation(s)
- Ravi Dhital
- Center for Vaccines and Immunity, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus OH
| | - Kaitlyn Flint
- Center for Vaccines and Immunity, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus OH
| | - Irina Kaptsan
- Center for Vaccines and Immunity, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus OH
| | - Shweta Hegde
- Center for Vaccines and Immunity, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus OH
| | - Reem Daloul
- Division of Transplant Nephrology, Comprehensive Transplant Center, The Ohio State University, Columbus OH
| | - Masako Shimamura
- Center for Vaccines and Immunity, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus OH
- Division of Pediatric Infectious Diseases, Department of Pediatrics, The Ohio State University, Columbus OH
| |
Collapse
|
66
|
Li Y, Xiao J, Li C, Yang M. Memory inflation: Beyond the acute phase of viral infection. Cell Prolif 2024; 57:e13705. [PMID: 38992867 PMCID: PMC11628752 DOI: 10.1111/cpr.13705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/09/2024] [Accepted: 06/14/2024] [Indexed: 07/13/2024] Open
Abstract
Memory inflation is confirmed as the most commonly dysregulation of host immunity with antigen-independent manner in mammals after viral infection. By generating large numbers of effector/memory and terminal differentiated effector memory CD8+ T cells with diminished naïve subsets, memory inflation is believed to play critical roles in connecting the viral infection and the onset of multiple diseases. Here, we reviewed the current understanding of memory inflated CD8+ T cells in their distinct phenotypic features that different from exhausted subsets; the intrinsic and extrinsic roles in regulating the formation of memory inflation; and the key proteins in maintaining the expansion and proliferation of inflationary populations. More importantly, based on the evidences from both clinic and animal models, we summarized the potential mechanisms of memory inflation to trigger autoimmune neuropathies, such as Guillain-Barré syndrome and multiple sclerosis; the correlations of memory inflation between tumorigenesis and resistance of tumour immunotherapies; as well as the effects of memory inflation to facilitate vascular disease progression. To sum up, better understanding of memory inflation could provide us an opportunity to beyond the acute phase of viral infection, and shed a light on the long-term influences of CD8+ T cell heterogeneity in dampen host immune homeostasis.
Collapse
Affiliation(s)
- Yanfei Li
- School of Basic Medical SciencesChengdu University of Traditional Chinese MedicineChengduChina
| | - Jie Xiao
- Centre for Translational Research in Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Chen Li
- Centre for Translational Research in Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Mu Yang
- School of Basic Medical SciencesChengdu University of Traditional Chinese MedicineChengduChina
- Centre for Translational Research in Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| |
Collapse
|
67
|
Ossami Saidy RR, Eurich F, Globke B, Schöning W, Öllinger R, Raschzok N, Pratschke J, Eurich D, Dittrich L, Dobrindt EM. The Association Between Cytomegalovirus Infection and Kidney Damage in the Liver Transplant Setting. Viruses 2024; 16:1830. [PMID: 39772140 PMCID: PMC11680441 DOI: 10.3390/v16121830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 01/11/2025] Open
Abstract
INTRODUCTION The development of chronic kidney disease (CKD) is a common and significant complication, contributing to morbidity after liver transplantation (LT). Cytomegalovirus (CMV) infection is common in the overall population, and relevant reinfection after LT may occur. CMV-associated kidney damage has been discussed, but the clinical significance on CKD development after LT remains unclear. METHODS A total of 745 patients who underwent LT between 2006 and 2017 were included in this retrospective analysis. Clinical data, as well as laboratory parameters, were analyzed. Univariate and multivariate analysis were performed. RESULTS The univariate analysis revealed significantly impaired estimated glomerular filtration rates (eGFRs) in patients with histories of CMV infection (81.4 (8-137) mL/min vs. 90.0 (5-147) mL/min; p = 0.004). This effect was confirmed in the multivariate analysis. Post-LT, eGFR was impaired in patients with CMV (re)infection at 6, 12, 36, and 60 months, 10 years, and 15 years after LT. Immunosuppressive levels were comparable between groups. Overall survival was negatively affected by CMV infection (p = 0.001). DISCUSSION A clinically significant detrimental impact of CMV infection on renal function was observed, that could individualize clinical risk evaluation prior and after LT further. However, the pathophysiological mechanisms behind this observation are not yet understood.
Collapse
Affiliation(s)
- Ramin Raul Ossami Saidy
- Department of Surgery, Campus Virchow Klinikum and Campus Charité Mitte, Charité–Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (F.E.); (B.G.); (W.S.); (R.Ö.); (N.R.); (J.P.); (D.E.); (L.D.); (E.M.D.)
| | - Franziska Eurich
- Department of Surgery, Campus Virchow Klinikum and Campus Charité Mitte, Charité–Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (F.E.); (B.G.); (W.S.); (R.Ö.); (N.R.); (J.P.); (D.E.); (L.D.); (E.M.D.)
| | - Brigitta Globke
- Department of Surgery, Campus Virchow Klinikum and Campus Charité Mitte, Charité–Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (F.E.); (B.G.); (W.S.); (R.Ö.); (N.R.); (J.P.); (D.E.); (L.D.); (E.M.D.)
- Berlin Institute of Health at Charité—Universitätsmedizin Berlin, BIH Academy, Clinician Scientist Program, 10117 Berlin, Germany
| | - Wenzel Schöning
- Department of Surgery, Campus Virchow Klinikum and Campus Charité Mitte, Charité–Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (F.E.); (B.G.); (W.S.); (R.Ö.); (N.R.); (J.P.); (D.E.); (L.D.); (E.M.D.)
| | - Robert Öllinger
- Department of Surgery, Campus Virchow Klinikum and Campus Charité Mitte, Charité–Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (F.E.); (B.G.); (W.S.); (R.Ö.); (N.R.); (J.P.); (D.E.); (L.D.); (E.M.D.)
| | - Nathanael Raschzok
- Department of Surgery, Campus Virchow Klinikum and Campus Charité Mitte, Charité–Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (F.E.); (B.G.); (W.S.); (R.Ö.); (N.R.); (J.P.); (D.E.); (L.D.); (E.M.D.)
- Berlin Institute of Health at Charité—Universitätsmedizin Berlin, BIH Academy, Clinician Scientist Program, 10117 Berlin, Germany
| | - Johann Pratschke
- Department of Surgery, Campus Virchow Klinikum and Campus Charité Mitte, Charité–Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (F.E.); (B.G.); (W.S.); (R.Ö.); (N.R.); (J.P.); (D.E.); (L.D.); (E.M.D.)
| | - Dennis Eurich
- Department of Surgery, Campus Virchow Klinikum and Campus Charité Mitte, Charité–Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (F.E.); (B.G.); (W.S.); (R.Ö.); (N.R.); (J.P.); (D.E.); (L.D.); (E.M.D.)
| | - Luca Dittrich
- Department of Surgery, Campus Virchow Klinikum and Campus Charité Mitte, Charité–Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (F.E.); (B.G.); (W.S.); (R.Ö.); (N.R.); (J.P.); (D.E.); (L.D.); (E.M.D.)
| | - Eva Maria Dobrindt
- Department of Surgery, Campus Virchow Klinikum and Campus Charité Mitte, Charité–Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (F.E.); (B.G.); (W.S.); (R.Ö.); (N.R.); (J.P.); (D.E.); (L.D.); (E.M.D.)
| |
Collapse
|
68
|
Fang L, Wang Z, Zhao J, Wu X, Wang S, Gao H, Wu D. Association between human herpesviruses infections and childhood neurodevelopmental disorders: insights from two-sample mendelian randomization analyses and systematic review with meta-analysis. Ital J Pediatr 2024; 50:248. [PMID: 39568007 PMCID: PMC11580506 DOI: 10.1186/s13052-024-01820-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 11/10/2024] [Indexed: 11/22/2024] Open
Abstract
BACKGROUND The potential roles of viral infections in neurodevelopmental disorders (NDDs) have been suggested based on previous studies. Given the high prevalence of human herpesviruses (HHVs), the associations between HHVs infection and the risk of NDDs warrant explored. METHODS Our study employs a two-sample Mendelian randomization (MR) analysis and systematic review with meta-analysis to investigate whether genetically predicted HHVs infection are linked to three main childhood NDDs-autism spectrum disorder (ASD), attention deficit hyperactivity disorder (ADHD), and Tourette syndrome (TS). We utilized genetic variants associated with HHV infections in genome-wide association study (GWAS) summary datasets of European populations to establish instrumental variables and statistics for three NDDs obtained from Psychiatric Genomics Consortium. MR analysis was performed using inverse-variance weighted, MR Egger, weighted median, simple median, weighted mode, and MR-PRESSO. In addition, publications associating HHVs infection with three NDDs were systematically searched using PubMed, Web of Science, and three Chinese databases for meta-analyses. RESULTS The MR results found no evidence to support a link between genetically predicted HHVs infection and the risk of NDDs based on existing datasets. Twenty-seven observational studies on children with HHVs infection and NDDs were considered eligible. Meta-analysis showed that cytomegalovirus and HHV-6 infection were related with ASD, while Epstein-Barr virus and cytomegalovirus infection were associated with TD in Chinese population. CONCLUSIONS These results contribute to a comprehensive understanding of the possibilities underlying HHV infections in affecting childhood NDDs. Further research is necessary to include larger and more robust statistics of HHV infections and NDDs. TRIAL REGISTRATION This systematic review was registered at PROSPERO as CRD42024554169. Retrospectively registered 26 July 2024.
Collapse
Affiliation(s)
- Liwei Fang
- Pediatric Neurorehabilitation Center, Pediatric Department, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zuojun Wang
- Pediatric Neurorehabilitation Center, Pediatric Department, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jingyi Zhao
- Pediatric Neurorehabilitation Center, Pediatric Department, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xun Wu
- Pediatric Neurorehabilitation Center, Pediatric Department, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shunxin Wang
- Pediatric Neurorehabilitation Center, Pediatric Department, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hui Gao
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui, China.
| | - De Wu
- Pediatric Neurorehabilitation Center, Pediatric Department, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
69
|
Semmes EC, Nettere DR, Nelson AN, Hurst JH, Cain DW, Burt TD, Kurtzberg J, Reeves RK, Coyne CB, Fouda GG, Pollara J, Permar SR, Walsh KM. In utero human cytomegalovirus infection expands NK-like FcγRIII+CD8+ T cells that mediate Fc antibody functions. J Clin Invest 2024; 135:e181342. [PMID: 39531313 PMCID: PMC11684805 DOI: 10.1172/jci181342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
Human cytomegalovirus (HCMV) profoundly impacts host T and NK cells across the lifespan, yet how this common congenital infection modulates developing fetal immune cell compartments remains underexplored. Using cord blood from neonates with and without congenital HCMV (cCMV) infection, we identify an expansion of Fcγ receptor III-expressing (FcγRIII-expressing) CD8+ T cells following HCMV exposure in utero. Most FcγRIII+CD8+ T cells express the canonical αβ T cell receptor (TCR), but a proportion express noncanonical γδ TCR. FcγRIII+CD8+ T cells are highly differentiated and have increased expression of NK cell markers and cytolytic molecules. Transcriptional analysis reveals FcγRIII+CD8+ T cells upregulate T-bet and downregulate BCL11B, known transcription factors that govern T/NK cell fate. We show that FcγRIII+CD8+ T cells mediate antibody-dependent IFN-γ production and degranulation against IgG-opsonized target cells, similar to NK cell antibody-dependent cellular cytotoxicity (ADCC). FcγRIII+CD8+ T cell Fc effector functions were further enhanced by IL-15, as has been observed in neonatal NK cells. Our study reveals that FcγRIII+CD8+ T cells elicited in utero by HCMV infection can execute Fc-mediated effector functions bridging cellular and humoral immunity and may be a promising target for antibody-based therapeutics and vaccination in early life.
Collapse
Affiliation(s)
- Eleanor C. Semmes
- Boston Children’s Hospital/Boston Medical Center, Boston, Massachusetts, USA
- Medical Scientist Training Program, and
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Danielle R. Nettere
- Medical Scientist Training Program, and
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Ashley N. Nelson
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Jillian H. Hurst
- Children’s Health and Discovery Initiative
- Division of Infectious Diseases, and
| | - Derek W. Cain
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Trevor D. Burt
- Children’s Health and Discovery Initiative
- Division of Neonatology, Department of Pediatrics, Duke University, Durham, North Carolina, USA
| | - Joanne Kurtzberg
- Children’s Health and Discovery Initiative
- Carolinas Cord Blood Bank, Marcus Center for Cellular Cures, Durham, North Carolina, USA
| | - R. Keith Reeves
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
- Center for Human Systems Immunology, and
- Department of Integrative Immunobiology, Duke University, Durham, North Carolina, USA
| | - Carolyn B. Coyne
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
- Department of Integrative Immunobiology, Duke University, Durham, North Carolina, USA
| | - Genevieve G. Fouda
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
- Children’s Health and Discovery Initiative
- Department of Pediatrics, Weill Cornell Medicine, New York City, New York, USA
| | - Justin Pollara
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Sallie R. Permar
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
- Children’s Health and Discovery Initiative
- Division of Infectious Diseases, and
- Department of Pediatrics, Weill Cornell Medicine, New York City, New York, USA
| | - Kyle M. Walsh
- Children’s Health and Discovery Initiative
- Department of Neurosurgery, Duke University, Durham, North Carolina, USA
| |
Collapse
|
70
|
Farrer TJ, Moore JD, Chase M, Gale SD, Hedges DW. Infectious Disease as a Modifiable Risk Factor for Dementia: A Narrative Review. Pathogens 2024; 13:974. [PMID: 39599527 PMCID: PMC11597442 DOI: 10.3390/pathogens13110974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/31/2024] [Accepted: 11/05/2024] [Indexed: 11/29/2024] Open
Abstract
This narrative review examines infectious diseases as modifiable risk factors for dementia, particularly in the context of an aging global population. As the prevalence of Alzheimer's disease and related dementias is expected to rise, prevention has become increasingly important due to the limited efficacy of current treatments. Emerging evidence links specific infectious diseases to increased dementia risk, possibly through mechanisms like neuroinflammation and disruption to normal cell function. Here, we review findings on how viral and bacterial infections contribute to dementia and explore potentially preventive measures, including vaccinations and antiviral treatments. Studies indicate that vaccinations against influenza, herpes zoster, and hepatitis, as well as antiviral treatments targeting human herpesvirus, could reduce the incidence of dementia. Additionally, non-pharmaceutical interventions during pandemics and in long-term care settings are highlighted as effective strategies for reducing the spread of infectious diseases, potentially lowering dementia risk. Putative mechanisms underlying the protective effects of these interventions suggest that reducing systemic inflammation may be important to their efficacy. While the currently available evidence suggests at best an association between some infectious diseases and dementia, this narrative review emphasizes the need to incorporate infectious disease prevention into broader public health strategies to potentially mitigate the growing burden of dementia. Further research is required to explore these preventive measures across diverse populations and to deepen our understanding of the biological mechanisms involved.
Collapse
Affiliation(s)
- Thomas J. Farrer
- Idaho WWAMI Medical Education Program, University of Idaho, Moscow, ID 83844, USA;
| | - Jonathan D. Moore
- Idaho WWAMI Medical Education Program, University of Idaho, Moscow, ID 83844, USA;
| | - Morgan Chase
- The Neuroscience Center, Brigham Young University, Provo, UT 84602, USA; (M.C.); (S.D.G.); (D.W.H.)
| | - Shawn D. Gale
- The Neuroscience Center, Brigham Young University, Provo, UT 84602, USA; (M.C.); (S.D.G.); (D.W.H.)
- The Department of Psychology, Brigham Young University, Provo, UT 84602, USA
| | - Dawson W. Hedges
- The Neuroscience Center, Brigham Young University, Provo, UT 84602, USA; (M.C.); (S.D.G.); (D.W.H.)
- The Department of Psychology, Brigham Young University, Provo, UT 84602, USA
| |
Collapse
|
71
|
Brochu HN, Smith E, Jeong S, Carlson M, Hansen SG, Tisoncik-Go J, Law L, Picker LJ, Gale M, Peng X. Pre-challenge gut microbial signature predicts RhCMV/SIV vaccine efficacy in rhesus macaques. Microbiol Spectr 2024; 12:e0128524. [PMID: 39345211 PMCID: PMC11537114 DOI: 10.1128/spectrum.01285-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 08/21/2024] [Indexed: 10/01/2024] Open
Abstract
Rhesus cytomegalovirus expressing simian immunodeficiency virus (RhCMV/SIV) vaccines protect ~59% of vaccinated rhesus macaques against repeated limiting-dose intra-rectal exposure with highly pathogenic SIVmac239M, but the exact mechanism responsible for the vaccine efficacy is unknown. It is becoming evident that complex interactions exist between gut microbiota and the host immune system. Here, we aimed to investigate if the rhesus gut microbiome impacts RhCMV/SIV vaccine-induced protection. Three groups of 15 rhesus macaques naturally pre-exposed to RhCMV were vaccinated with RhCMV/SIV vaccines. Rectal swabs were collected longitudinally both before SIV challenge (after vaccination) and post-challenge and were profiled using 16S rRNA based microbiome analysis. We identified ~2,400 16S rRNA amplicon sequence variants (ASVs), representing potential bacterial species/strains. Global gut microbial profiles were strongly associated with each of the three vaccination groups, and all animals tended to maintain consistent profiles throughout the pre-challenge phase. Despite vaccination group differences, by using newly developed compositional data analysis techniques, we identified a common gut microbial signature predictive of vaccine protection outcome across the three vaccination groups. Part of this microbial signature persisted even after SIV challenge. We also observed a strong correlation between this microbial signature and an early signature derived from whole blood transcriptomes in the same animals. Our findings indicate that changes in gut microbiomes are associated with RhCMV/SIV vaccine-induced protection and early host response to vaccination in rhesus macaques.IMPORTANCEThe human immunodeficiency virus (HIV) has infected millions of people worldwide. Unfortunately, still there is no vaccine that can prevent or treat HIV infection. A promising pre-clinical HIV vaccine based on rhesus cytomegalovirus (RhCMV) expressing simian immunodeficiency virus (SIV) antigens (RhCMV/SIV) provides sustained, durable protection against SIV challenge in ~59% of vaccinated rhesus macaques. There is an urgent need to understand the cause of this protection vs non-protection outcome. In this study, we profiled the gut microbiomes of 45 RhCMV/SIV vaccinated rhesus macaques and identified gut microbial signatures that were predictive of RhCMV/SIV vaccination groups and vaccine protection outcomes. These vaccine protection-associated microbial features were significantly correlated with early vaccine-induced host immune signatures in whole blood from the same animals. These findings show that the gut microbiome may be involved in RhCMV/SIV vaccine-induced protection, warranting further research into the impact of the gut microbiome in human vaccine trials.
Collapse
Affiliation(s)
- Hayden N. Brochu
- Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina, USA
- Bioinformatics Graduate Program, North Carolina State University, Raleigh, North Carolina, USA
| | - Elise Smith
- Department of Immunology, University of Washington, Seattle, Washington, USA
| | - Sangmi Jeong
- Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina, USA
- Bioinformatics Graduate Program, North Carolina State University, Raleigh, North Carolina, USA
| | - Michelle Carlson
- Department of Immunology, University of Washington, Seattle, Washington, USA
| | - Scott G. Hansen
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Jennifer Tisoncik-Go
- Department of Immunology, University of Washington, Seattle, Washington, USA
- Center for Innate Immunity and Immune Disease, University of Washington, Seattle, Washington, USA
| | - Lynn Law
- Department of Immunology, University of Washington, Seattle, Washington, USA
- Center for Innate Immunity and Immune Disease, University of Washington, Seattle, Washington, USA
| | - Louis J. Picker
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Michael Gale
- Department of Immunology, University of Washington, Seattle, Washington, USA
- Center for Innate Immunity and Immune Disease, University of Washington, Seattle, Washington, USA
- Washington National Primate Research Center, University of Washington, Seattle, Washington, USA
| | - Xinxia Peng
- Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina, USA
- Bioinformatics Graduate Program, North Carolina State University, Raleigh, North Carolina, USA
- Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
72
|
Lai S, Wei W, Yang S, Wu Y, Shi M, Meng S, Tao X, Chen S, Chen R, Su J, Yuan Z, Ye L, Liang H, Xie Z, Jiang J. Machine learning-driven in-hospital mortality prediction in HIV/AIDS patients with Cytomegalovirus infection: a single-centred retrospective study. J Med Microbiol 2024; 73. [PMID: 39606806 DOI: 10.1099/jmm.0.001935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2024] Open
Abstract
Introduction. Cytomegalovirus (CMV) is a widely disseminated betaherpesvirus that typically induces latant infections. In immunocompromised populations, especially transplant and HIV-infected patients, CMV infection increases in-hospital mortality.Gap statement. Although machine learning models have been widely used in clinical diagnosis and prognosis prediction, reports on machine learning model predictions for the in-hospital mortality of HIV/AIDS patients with CMV infection have not been reported.Aim. Analyze the general gemographic and clinical characteristics of HIV/AIDS patients with CMV infection and identify the factors affecting the prognosis of this population, which will help to reduce their in-hospital mortality.Methods. Hospitalized HIV/AIDS patients with CMV infection were recruited from the Fourth People's Hospital of Nanning, Guangxi, from 2012 to 2019. After dividing them into survival and death groups based on their in-hospital survival status, their general and clinical profiles were described. Following 1 : 3 propensity score matching to equalize baseline characteristics, three machine-learning models (Random Forest, Support Vector Machine and eXtreme Gradient Boosting) were deployed to forecast factors influencing prognosis. The SHapley Additive exPlanations tool explained the models.Results. A total of 1102 HIV/AIDS patients with CMV infection were analysed. There was no statistical difference in the general condition of the study subjects (P>0.05). Prevalent complications/coinfections included pneumonia (63.6%), tuberculosis (47.2%) and oral fungal infections (44.6%). There were significant differences between the groups in pneumonia, cryptococcosis and hypoproteinaemia (P<0.05). The differences in laboratory indicators between patients were also statistically significant (P<0.05). The three machine learning models demonstrated good performance, identifying primary predictors of mortality. Pneumonia, urea, indirect bilirubin and platelet distribution width exhibited positive associations with death, with higher levels correlating with an increased mortality risk. Conversely, CD4 T-cell count, CD8 T-cell count and platelet displayed negative correlations with mortality.Conclusions. HIV/AIDS patients with CMV infection exhibit distinctive clinical features impacting survival outcomes. Machine learning models accurately identify key influencing factors and predict mortality risk in this population, which appears to be essential to reducing in-hospital mortality.
Collapse
Affiliation(s)
- Shiyi Lai
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Wudi Wei
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, PR China
- Joint Laboratory for Emerging Infectious Diseases in China (Guangxi)-ASEAN, Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Shixiong Yang
- The Fourth People's Hospital of Nanning, Nanning, Guangxi, PR China
| | - Yuting Wu
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Minjuan Shi
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Sirun Meng
- The Fourth People's Hospital of Nanning, Nanning, Guangxi, PR China
| | - Xing Tao
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Shanshan Chen
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Rongfeng Chen
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, PR China
- Joint Laboratory for Emerging Infectious Diseases in China (Guangxi)-ASEAN, Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Jinming Su
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, PR China
- Joint Laboratory for Emerging Infectious Diseases in China (Guangxi)-ASEAN, Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Zongxiang Yuan
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Li Ye
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, PR China
- Joint Laboratory for Emerging Infectious Diseases in China (Guangxi)-ASEAN, Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Hao Liang
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, PR China
- Joint Laboratory for Emerging Infectious Diseases in China (Guangxi)-ASEAN, Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Zhiman Xie
- The Fourth People's Hospital of Nanning, Nanning, Guangxi, PR China
| | - Junjun Jiang
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, PR China
- Joint Laboratory for Emerging Infectious Diseases in China (Guangxi)-ASEAN, Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, PR China
| |
Collapse
|
73
|
Jun CH, Kim SH, Kim J, Kim KM, Lee JW, Kong SM, Kwak J, Kwan BS, Cho HK, Park KJ, Kim HT, Wi YM. Quantitative PCR for early detection of human cytomegalovirus end-organ disease in immunocompetent host: A retrospective single-center study. J Infect Public Health 2024; 17:102542. [PMID: 39299079 DOI: 10.1016/j.jiph.2024.102542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/04/2024] [Accepted: 09/12/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Tissue-invasive end-organ disease (EOD) caused by cytomegalovirus (CMV) is less frequently reported in immunocompetent patients compared to immunocompromised patients. In this study, we investigated the association between CMV viremia and CMV end-organ disease in immunocompetent patients. METHODS Adult patients (≥18 years old) with CMV viremia were screened from January 2010 to June 2022. The primary outcome was the presence of CMV EOD. Risk factors associated with CMV EOD were analyzed, and a receiver operating characteristic curve was plotted to determine the most accurate cutoff value of the CMV titer for the prediction of CMV EOD. RESULTS Among the 106 immunocompetent patients with CMV viremia, 31 exhibited CMV EOD. Gastrointestinal tract disease was the most common. The log10 value of the CMV titer was significantly associated with the occurrence of CMV EOD in immunocompetent patients with CMV viremia. The optimal cut-off CMV titer for the prediction of CMV EOD was 749 IU/mL. CONCLUSIONS Our study suggests the potential association between high CMV titers and the development of CMV end-organ diseases and describes the diagnostic performance and utility of quantitative PCR as a surrogate marker for predicting the occurrence of CMV EOD in immunocompetent patients.
Collapse
Affiliation(s)
- Cheon-Hoo Jun
- Department of Infectious Diseases, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Republic of Korea
| | - Si-Ho Kim
- Department of Infectious Diseases, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Republic of Korea
| | - Junyoung Kim
- Department of Internal Medicine, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Republic of Korea
| | - Kwang Min Kim
- Department of Internal Medicine, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Republic of Korea
| | - Jung Won Lee
- Department of Internal Medicine, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Republic of Korea
| | - Sung Min Kong
- Department of Internal Medicine, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Republic of Korea
| | - Jiyeong Kwak
- Department of Internal Medicine, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Republic of Korea
| | - Byung Soo Kwan
- Department of Internal Medicine, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Republic of Korea
| | - Hyun Kyu Cho
- Department of Internal Medicine, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Republic of Korea
| | - Kyoung-Jin Park
- Department of Laboratory Medicine and Genetics, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Republic of Korea
| | - Hyoung Tae Kim
- Department of Laboratory Medicine and Genetics, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Republic of Korea
| | - Yu Mi Wi
- Department of Infectious Diseases, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Republic of Korea.
| |
Collapse
|
74
|
Mukhiya R, Fleischmann WA, Loughland JR, Chan JA, de Labastida Rivera F, Andrew D, Beeson JG, McCarthy JS, Barber BE, Lopez JA, Engwerda C, Thomson-Luque R, Boyle MJ. Heterogeneity of the human immune response to malaria infection and vaccination driven by latent cytomegalovirus infection. EBioMedicine 2024; 109:105419. [PMID: 39490199 PMCID: PMC11576503 DOI: 10.1016/j.ebiom.2024.105419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 11/05/2024] Open
Abstract
BACKGROUND Human immune responses to infection and vaccination are heterogenous, driven by multiple factors including genetics, environmental exposures and personal infection histories. For malaria caused by Plasmodium falciparum parasites, host factors that impact on humoral immunity are poorly understood. METHODS We investigated the role of latent cytomegalovirus (CMV) on the host immune response to malaria using samples obtained from individuals in previously conducted Phase 1 trials of blood stage P. falciparum Controlled Human Malaria Infection (CHMI) and in a MSP1 vaccine clinical trial. Induced antibody and functions of antibodies, as well as CD4 T cell responses were quantified. FINDINGS CMV seropositivity was associated with reduced induction of parasite specific antibodies following malaria infection and vaccination. During infection, reduced antibody induction was associated with modifications to the T -follicular helper (Tfh) cell compartment. CMV seropositivity was associated with a skew towards Tfh1 cell subsets before and after malaria infection, and reduced activation of Tfh2 cells. Protective Tfh2 cell activation was only associated with antibody development in individuals who were CMV seronegative, and a higher proportion of Tfh1 cells was associated with lower antibody development in individuals who were CMV seropositive. During MSP1 vaccination, reduced antibody induction in individuals who were CMV seropositive was associated with CD4 T cell expression of terminal differentiation marker CD57. INTERPRETATION These findings suggest that CMV seropositivity may be negatively associated with malaria antibody development. Further studies in larger cohorts, particularly in malaria endemic regions are required to investigate whether CMV infection may modify immunity to malaria gained during infection or vaccination in children. FUNDING Work was funded by National Health and Medical Research Council of Australia, CSL Australia and Snow Medical Foundation. Funders had no role in data generation, writing of manuscript of decision to submit for publication.
Collapse
Affiliation(s)
- Reena Mukhiya
- Burnet Institute, Melbourne, Australia; School of Environmental Sciences, Griffith University, Brisbane, Australia; QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Wim A Fleischmann
- Center for Infectious Diseases, Virology, Heidelberg University, Medical Faculty, University Hospital Heidelberg, Germany
| | - Jessica R Loughland
- Burnet Institute, Melbourne, Australia; QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Jo-Anne Chan
- Burnet Institute, Melbourne, Australia; Department of Infectious Diseases, University of Melbourne, Australia; Department of Microbiology and School of Translational Medicine, Monash University, Australia
| | | | - Dean Andrew
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - James G Beeson
- Burnet Institute, Melbourne, Australia; Department of Infectious Diseases, University of Melbourne, Australia; Department of Microbiology and School of Translational Medicine, Monash University, Australia
| | - James S McCarthy
- QIMR Berghofer Medical Research Institute, Brisbane, Australia; Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | | | - J Alejandro Lopez
- School of Environmental Sciences, Griffith University, Brisbane, Australia; QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Christian Engwerda
- School of Environmental Sciences, Griffith University, Brisbane, Australia; QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Richard Thomson-Luque
- Sumaya-Biotech GmbH & Co. KG, Germany; Centre for Infectious Diseases, Parasitology, Heidelberg University, Medical Faculty, University Hospital Heidelberg, Germany
| | - Michelle J Boyle
- Burnet Institute, Melbourne, Australia; School of Environmental Sciences, Griffith University, Brisbane, Australia; QIMR Berghofer Medical Research Institute, Brisbane, Australia; Department of Infectious Diseases, University of Melbourne, Australia; Department of Microbiology and School of Translational Medicine, Monash University, Australia.
| |
Collapse
|
75
|
Al Beloushi M, Saleh H, Ahmed B, Konje JC. Congenital and Perinatal Viral Infections: Consequences for the Mother and Fetus. Viruses 2024; 16:1698. [PMID: 39599813 PMCID: PMC11599085 DOI: 10.3390/v16111698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 10/13/2024] [Accepted: 10/26/2024] [Indexed: 11/29/2024] Open
Abstract
Viruses are the most common congenital infections in humans and an important cause of foetal malformations, neonatal morbidity, and mortality. The effects of these infections, which are transmitted in utero (transplacentally), during childbirth or in the puerperium depend on the timing of the infections. These vary from miscarriages (usually with infections in very early pregnancy), congenital malformations (when the infections occur during organogenesis) and morbidity (with infections occurring late in pregnancy, during childbirth or after delivery). The most common of these viruses are cytomegalovirus, hepatitis, herpes simplex type-2, parvovirus B19, rubella, varicella zoster and zika viruses. There are currently very few efficacious antiviral agents licensed for use in pregnancy. For most of these infections, therefore, prevention is mainly by vaccination (where there is a vaccine). The administration of immunoglobulins to those exposed to the virus to offer passive immunity or appropriate measures to avoid being infected would be options to minimise the infections and their consequences. In this review, we discuss some of the congenital and perinatal infections and their consequences on both the mother and fetus and their management focusing mainly on prevention.
Collapse
Affiliation(s)
- Mariam Al Beloushi
- Women’s Wellness and Research Centre Hamad Medical Corporation, Doha P.O. Box 3050, Qatar; (M.A.B.); (H.S.)
- Department of Obstetrics and Gynaecology, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Huda Saleh
- Women’s Wellness and Research Centre Hamad Medical Corporation, Doha P.O. Box 3050, Qatar; (M.A.B.); (H.S.)
- Department of Obstetrics and Gynaecology, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Badreldeen Ahmed
- Department of Obstetrics and Gynaecology, Qatar University, Doha P.O. Box 2713, Qatar;
- Feto Maternal Centre, Al Markhiya Doha, Doha P.O. Box 34181, Qatar
- Department of Obstetrics and Gynaecology Weill Cornell Medicine, Doha P.O. Box 24144, Qatar
| | - Justin C. Konje
- Feto Maternal Centre, Al Markhiya Doha, Doha P.O. Box 34181, Qatar
- Department of Obstetrics and Gynaecology Weill Cornell Medicine, Doha P.O. Box 24144, Qatar
- Department of Health Sciences, University of Leicester, P.O. Box 7717, Leicester LE2 7LX, UK
| |
Collapse
|
76
|
Blanco R, Muñoz JP. HPV and HCMV in Cervical Cancer: A Review of Their Co-Occurrence in Premalignant and Malignant Lesions. Viruses 2024; 16:1699. [PMID: 39599814 PMCID: PMC11599080 DOI: 10.3390/v16111699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/24/2024] [Accepted: 10/26/2024] [Indexed: 11/29/2024] Open
Abstract
Cervical cancer remains a significant global health concern, particularly in low- and middle-income countries. While persistent infection with high-risk human papillomavirus (HR-HPV) is essential for cervical cancer development, it is not sufficient on its own, suggesting the involvement of additional cofactors. The human cytomegalovirus (HCMV) is a widespread β-herpesvirus known for its ability to establish lifelong latency and reactivate under certain conditions, often contributing to chronic inflammation and immune modulation. Emerging evidence suggests that HCMV may play a role in various cancers, including cervical cancer, through its potential to influence oncogenic pathways and disrupt host immune responses. This review explores clinical evidence regarding the co-presence of HR-HPV and HCMV in premalignant lesions and cervical cancer. The literature reviewed indicates that HCMV is frequently detected in cervical lesions, particularly in those co-infected with HPV, suggesting a potential synergistic interaction that could enhance HPV's oncogenic effects, thereby facilitating the progression from low-grade squamous intraepithelial lesions (LSIL) to high-grade squamous intraepithelial lesions (HSIL) and invasive cancer. Although the precise molecular mechanisms were not thoroughly investigated in this review, the clinical evidence suggests the importance of considering HCMV alongside HPV in the management of cervical lesions. A better understanding of the interaction between HR-HPV and HCMV may lead to improved diagnostic, therapeutic, and preventive strategies for cervical cancer.
Collapse
Affiliation(s)
- Rancés Blanco
- Independent Researcher, Av. Vicuña Mackenna Poniente 6315, La Florida 8240000, Chile
| | - Juan P. Muñoz
- Laboratorio de Bioquímica, Departamento de Química, Facultad de Ciencias, Universidad de Tarapacá, Arica 1000007, Chile
| |
Collapse
|
77
|
Evans EF, Saraph A, Tokuyama M. Transactivation of Human Endogenous Retroviruses by Viruses. Viruses 2024; 16:1649. [PMID: 39599764 PMCID: PMC11599155 DOI: 10.3390/v16111649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/12/2024] [Accepted: 10/16/2024] [Indexed: 11/29/2024] Open
Abstract
Human endogenous retroviruses (HERVs) are remnants of ancient retroviral infections that are part the human genome and are normally silenced through epigenetic mechanisms. However, HERVs can be induced by various host and environmental factors, including viral infection, and transcriptionally active HERVs have been implicated in various physiological processes. In this review, we summarize mounting evidence of transactivation of HERVs by a wide range of DNA and RNA viruses. Though a mechanistic understanding of this phenomenon and the biological implications are still largely missing, the link between exogenous and endogenous viruses is intriguing. Considering the increasing recognition of the role of viral infections in disease, understanding these interactions provides novel insights into human health.
Collapse
Affiliation(s)
| | | | - Maria Tokuyama
- Department of Microbiology and Immunology, Life Sciences Institute, The University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| |
Collapse
|
78
|
Norris MJ, Henderson LA, Siddiquey MNA, Yin J, Yoo K, Brunel S, Saphire EO, Benedict CA, Kamil JP. A noncanonical glycoprotein H complex enhances cytomegalovirus entry. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.13.617647. [PMID: 39416215 PMCID: PMC11482907 DOI: 10.1101/2024.10.13.617647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Human cytomegalovirus (HCMV) causes severe birth defects, lifelong health complications, and $4 billion in annual costs in the United States alone. A major challenge in vaccine design is the incomplete understanding of the diverse protein complexes the virus uses to infect cells. In Herpesviridae, the gH/gL glycoprotein heterodimer is expected to be a basal element of virion cell entry machinery. For HCMV, gH/gL forms a "trimer" with gO and a "pentamer" with UL128, UL130, and UL131A, with each complex binding distinct receptors to enter varied cell types. Here, we reveal a third glycoprotein complex, abundant in HCMV virions, which significantly enhances infection of endothelial cells. In this "3-mer" complex, gH, without gL, associates with UL116 and UL141, an immunoevasin previously known to function in an intracellular role. Cryo-EM reveals the virion-surface 3-mer is structurally unique among Herpesviridae gH complexes, with gH-only scaffolding, UL141-mediated dimerization and a heavily glycosylated UL116 cap. Given that antibodies directed at gH and UL141 each can restrict HCMV replication, our work highlights this virion surface complex as a new target for vaccines and antiviral therapies.
Collapse
Affiliation(s)
- Michael J Norris
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA
- Present address (M.N.): Department of Biochemistry, University of Toronto, Toronto, ON Canada
| | - Lauren A Henderson
- Department of Microbiology and Immunology, Louisiana State University Health Shreveport, Shreveport, Shreveport, LA
- Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Shreveport, Shreveport, LA
| | - Mohammed N A Siddiquey
- Department of Microbiology and Immunology, Louisiana State University Health Shreveport, Shreveport, Shreveport, LA
| | - Jieyun Yin
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA
| | - Kwangsun Yoo
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA
| | - Simon Brunel
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA
| | - Erica Ollmann Saphire
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA
- Department of Medicine, University of California San Diego, La Jolla, CA
| | - Chris A Benedict
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA
| | - Jeremy P Kamil
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
79
|
Malouli D, Taher H, Mansouri M, Iyer RF, Reed J, Papen C, Schell JB, Beechwood T, Martinson T, Morrow D, Hughes CM, Gilbride RM, Randall K, Ford JC, Belica K, Ojha S, Sacha JB, Bimber BN, Hansen SG, Picker LJ, Früh K. Human cytomegalovirus UL18 prevents priming of MHC-E- and MHC-II-restricted CD8 + T cells. Sci Immunol 2024; 9:eadp5216. [PMID: 39392895 PMCID: PMC11797217 DOI: 10.1126/sciimmunol.adp5216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 09/19/2024] [Indexed: 10/13/2024]
Abstract
Rhesus cytomegalovirus (RhCMV) vectors elicit major histocompatibility complex (MHC)-E-restricted CD8+ T cells that stringently control simian immunodeficiency virus (SIV) in rhesus macaques. These responses require deletion of eight RhCMV chemokine-like open reading frames (ORFs) that are conserved in human cytomegalovirus (HCMV). To determine whether HCMV encodes additional, nonconserved inhibitors of unconventional T cell priming, we inserted 41 HCMV-specific ORFs into a chemokine-deficient strain (68-1 RhCMV). Monitoring of epitope recognition revealed that HCMV UL18 prevented unconventional T cell priming, resulting in MHC-Ia-targeted responses. UL18 is homologous to MHC-I but does not engage T cell receptors and, instead, binds with high affinity to inhibitory leukocyte immunoglobulin-like receptor-1 (LIR-1). UL18 lacking LIR-1 binding no longer interfered with MHC-E-restricted T cell stimulation by RhCMV-infected cells or the induction of unconventionally restricted T cells. Thus, LIR-1 binding needs to be deleted from UL18 of HCMV/HIV vaccines to allow for the induction of protective MHC-E-restricted T cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Scott G. Hansen
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Louis J. Picker
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Klaus Früh
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| |
Collapse
|
80
|
Xia J, Kantipudi S, Striebich CC, Henao-Martinez AF, Manoharan N, Palestine AG, Reddy AK. Cytomegalovirus chronic retinal necrosis with ganciclovir resistance: a case report. J Ophthalmic Inflamm Infect 2024; 14:50. [PMID: 39377839 PMCID: PMC11461363 DOI: 10.1186/s12348-024-00434-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 09/23/2024] [Indexed: 10/09/2024] Open
Abstract
BACKGROUND Cytomegalovirus (CMV) chronic retinal necrosis (CRN) is a rare viral retinal infection that occurs in mildly immunocompromised people. It shares some features with both acute retinal necrosis and CMV retinitis. It is typically treated with combination intravitreal and systemic ganciclovir. We discuss the management of a case of CMV CRN with ganciclovir resistance. CASE PRESENTATION An 80-year-old female presented with one month of blurry vision in the left eye. She was being treated with abatacept, methotrexate, and prednisone for rheumatoid arthritis. Examination revealed anterior chamber and vitreous cell along with peripheral retinal whitening. Fluorescein angiogram showed diffuse retinal non-perfusion. Aqueous fluid PCR testing returned positive for CMV. The retinitis was initially controlled with oral and intravitreal ganciclovir, but then recurred and progressed despite these therapies. Ganciclovir resistance was suspected and the patient was switched to intravitreal foscarnet injections, along with oral letermovir and leflunomide, which lead to resolution of the retinitis. The patient has now continued with letermovir and leflunomide for approximately 2.5 years without reactivation of the retinitis or need for further intravitreal anti-viral injections and with adequate control of her rheumatoid arthritis. CONCLUSION The incidence of CMV CRN may increase in the future as the use of non-cytotoxic immunosuppressive therapies that result in relatively mild immunosuppression also increases. Treatment with ganciclovir is effective but frequently leads to resistance, as in our case. In this situation, combination therapy with letermovir and leflunomide, particularly in the setting of rheumatoid arthritis where leflunomide can also have an anti-inflammatory effect, can be considered.
Collapse
Affiliation(s)
- Julia Xia
- Department of Ophthalmology, University of Colorado School of Medicine, 1675 Aurora Court, F731, Aurora, CO, 80045, USA
| | - Sanjana Kantipudi
- Department of Ophthalmology, University of Colorado School of Medicine, 1675 Aurora Court, F731, Aurora, CO, 80045, USA
| | - Christopher C Striebich
- Division of Rheumatology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Andrés F Henao-Martinez
- Division of Infectious Diseases, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Niranjan Manoharan
- Department of Ophthalmology, University of Colorado School of Medicine, 1675 Aurora Court, F731, Aurora, CO, 80045, USA
| | - Alan G Palestine
- Department of Ophthalmology, University of Colorado School of Medicine, 1675 Aurora Court, F731, Aurora, CO, 80045, USA
| | - Amit K Reddy
- Department of Ophthalmology, University of Colorado School of Medicine, 1675 Aurora Court, F731, Aurora, CO, 80045, USA.
| |
Collapse
|
81
|
Wei M, Zhang Y, Li Z, Liang Q, Cao T, Ma J. Epidemiological characteristics of three herpesviruses infections in children in Nanjing, China, from 2018 to 2023. Front Cell Infect Microbiol 2024; 14:1448533. [PMID: 39421641 PMCID: PMC11484055 DOI: 10.3389/fcimb.2024.1448533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 08/13/2024] [Indexed: 10/19/2024] Open
Abstract
Objective To evaluate the epidemiology characteristics of Herpes simplex virus type 2 (HSV-2), Epstein-Barr virus (EBV) and Cytomegalovirus (CMV) infection in children from January 2018 to December 2023, in Nanjing, China. Methods We conducted a retrospective analysis of 21,210, 49,494 and 32,457 outpatients and inpatients aged 1 day to 17 years who were subjected to the three herpesviruses (HSV-2, EBV, and CMV) nucleic acid testing from January 2018 to December 2023, respectively. Demographic information, laboratory findings, etc. were collected and analyzed. HSV-2, EBV and CMV nucleic acid testing were performed by real-time PCR. Results The total rate of detection of the three herpesviruses for all specimens was 0.32% (67/21,210), 14.99% (7419/49,494), and 8.88% (2881/32,457), respectively. A declining trend in the incidence of viral infections over the years was observed for the three herpesviruses (all P<0.05). The detection rate for HSV-2, EBV, and CMV was highest among patients aged 1-3 years, 3-7 years, and 28 days to 1 year, respectively (all P<0.05). The presence of HSV-2 and CMV infection did not exhibit a discernible seasonal pattern, whereas EBV typically demonstrated an elevation during the summer and autumn. Conclusion EBV and CMV were both prevalent among children in China, except for HSV-2. The annual prevalence of the three herpesviruses show decreasing trend from 2018 to 2023, and no difference in gender (except for EBV). EBV infections usually occur in the summer and autumn, whereas HSV-2 and CMV do not exhibit significant seasonality. The positivity rate of HSV-2 is highest in 1-3 years, EBV is highest in 3-7 years, and that of CMV is highest in 28 days to 1 year. Positive detection rates are higher in outpatients than in inpatients.
Collapse
Affiliation(s)
- Mingwei Wei
- NHC Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, Jiangsu, China
| | - Yang Zhang
- Department of Clinical Laboratory, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Zhibin Li
- Department of Immunology, Nanjing Medical University, Nanjing, China
| | - Qi Liang
- NHC Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, Jiangsu, China
| | - Tong Cao
- Department of Clinical Laboratory, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Jingjing Ma
- Department of Clinical Laboratory, Children’s Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
82
|
Balegamire SJ, Mâsse B, Audibert F, Lamarre V, Giguere Y, Forest JC, Boucoiran I. Association Between Maternal Cytomegalovirus Seropositivity, Preterm Birth, and Preeclampsia in Two Cohorts From Quebec, Canada: A Mediation Analysis. Am J Reprod Immunol 2024; 92:e13941. [PMID: 39436114 DOI: 10.1111/aji.13941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 08/14/2024] [Accepted: 09/23/2024] [Indexed: 10/23/2024] Open
Abstract
PROBLEM Preterm birth and preeclampsia significantly contribute to infant morbidity and mortality, posing critical public health concerns. Viral infections, particularly Cytomegalovirus (CMV), associated with chronic inflammation, may play a role in these adverse pregnancy outcomes. The contribution of CMV to preterm birth and preeclampsia requires further investigation. METHOD OF STUDY Data from 6048 pregnant women from two prospective Quebec cohorts, recruited between May 2005 and August 2012, were analyzed. First-trimester CMV serology was the exposure variable. Associations were assessed using multivariable logistic regression adjusted by inverse probability treatment weighting (IPTW) of propensity scores. Mediation analyses estimated the direct effect of maternal CMV serostatus on preterm birth, excluding mediation by preeclampsia. RESULTS Preterm birth and preeclampsia proportions were 5.1% (95% CI: 4.6-5.7) and 1.9% (95% CI: 1.6-2.3), respectively. Multivariable logistic regression adjusted by IPTW showed associations between CMV seropositivity and preterm birth (OR 1.20, 95% CI: 1.02-1.41) and CMV seropositivity and preeclampsia (OR 1.41, 95% CI: 1.08-1.84). Mediation analysis indicated that 97% of the total effect of CMV seropositivity on preterm birth is direct, with the remaining 3% mediated by preeclampsia. CONCLUSIONS CMV seropositivity appears to be a risk factor for both preterm birth and preeclampsia. The effect of maternal CMV seropositivity on preterm birth is primarily direct, not mediated by preeclampsia. Future studies should explore the impact of preventive measures against CMV infection on the incidence of preterm delivery and preeclampsia.
Collapse
Affiliation(s)
- Safari Joseph Balegamire
- Department of Social and Preventive Medicine, École de Santé Publique de Université de Montréal, Montreal, Quebec, Canada
- Women and Children's Infectious Diseases Center, CHU Sainte-Justine Research Center, Montreal, Canada
| | - Benoît Mâsse
- Department of Social and Preventive Medicine, École de Santé Publique de Université de Montréal, Montreal, Quebec, Canada
- Applied Clinical Research Unit, CHU Sainte Justine Research Center, Montreal, Canada
| | - François Audibert
- Department of Obstetrics and Gynecology, Division of Maternofetal Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Valerie Lamarre
- Department of Obstetrics and Gynecology, Division of Maternofetal Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Yves Giguere
- CHU de Québec-Université Laval Research Center, Quebec City, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Jean-Claude Forest
- CHU de Québec-Université Laval Research Center, Quebec City, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Isabelle Boucoiran
- Department of Social and Preventive Medicine, École de Santé Publique de Université de Montréal, Montreal, Quebec, Canada
- Women and Children's Infectious Diseases Center, CHU Sainte-Justine Research Center, Montreal, Canada
- Department of Obstetrics and Gynecology, Division of Maternofetal Medicine, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
83
|
Correale J, Solomon AJ, Cohen JA, Banwell BL, Gracia F, Gyang TV, de Bedoya FHD, Harnegie MP, Hemmer B, Jacob A, Kim HJ, Marrie RA, Mateen FJ, Newsome SD, Pandit L, Prayoonwiwat N, Sahraian MA, Sato DK, Saylor D, Shi FD, Siva A, Tan K, Viswanathan S, Wattjes MP, Weinshenker B, Yamout B, Fujihara K. Differential diagnosis of suspected multiple sclerosis: global health considerations. Lancet Neurol 2024; 23:1035-1049. [PMID: 39304243 DOI: 10.1016/s1474-4422(24)00256-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 05/14/2024] [Accepted: 06/04/2024] [Indexed: 09/22/2024]
Abstract
The differential diagnosis of multiple sclerosis can present specific challenges in patients from Latin America, Africa, the Middle East, eastern Europe, southeast Asia, and the Western Pacific. In these areas, environmental factors, genetic background, and access to medical care can differ substantially from those in North America and western Europe, where multiple sclerosis is most common. Furthermore, multiple sclerosis diagnostic criteria have been developed primarily using data from North America and western Europe. Although some diagnoses mistaken for multiple sclerosis are common regardless of location, a comprehensive approach to the differential diagnosis of multiple sclerosis in Latin America, Africa, the Middle East, eastern Europe, southeast Asia, and the Western Pacific regions requires special consideration of diseases that are prevalent in those locations. A collaborative effort has therefore assessed global differences in multiple sclerosis differential diagnoses and proposed recommendations for evaluating patients with suspected multiple sclerosis in regions beyond North America and western Europe.
Collapse
Affiliation(s)
- Jorge Correale
- Department of Neurology, Fleni, Buenos Aires, Argentina; Institute of Biological Chemistry and Biophysics, CONICET/University of Buenos Aires, Buenos Aires, Argentina.
| | - Andrew J Solomon
- Department of Neurological Sciences, Larner College of Medicine at the University of Vermont, Burlington, VT, USA
| | - Jeffrey A Cohen
- Department of Neurology, Mellen Center for Multiple Sclerosis Treatment and Research, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Brenda L Banwell
- Division of Child Neurology, Department of Pediatrics, The Children's Hospital of Philadelphia, Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Fernando Gracia
- Hospital Santo Tomás, Panama City, Panama; Universidad Interamericana de Panamá, School of Medicine, Panama City, Panama
| | - Tirisham V Gyang
- Department of Neurology, The Ohio State University, Columbus, Ohio, USA
| | | | - Mary P Harnegie
- Cleveland Clinic Libraries, Cleveland Clinic, Cleveland, Ohio, USA
| | - Bernhard Hemmer
- Department of Neurology, Klinikum Rechts der Isar, School of Medicine and Health, Technical University of Munich, Munich Cluster for Systems Neurology, Munich, Germany
| | - Anu Jacob
- Cleveland Clinic, Abu Dhabi, United Arab Emirates
| | - Ho Jin Kim
- Department of Neurology, National Cancer Center, Goyang, South Korea
| | - Ruth Ann Marrie
- Departments of Internal Medicine and Community Health Sciences, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Farrah J Mateen
- Department of Neurology, Massachusetts General Hospital, Harvard University, Boston, USA
| | - Scott D Newsome
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lekha Pandit
- Center for Advanced Neurological Research, KS Hedge Medical Academy, Nitte University, Mangalore, India
| | - Naraporn Prayoonwiwat
- Division of Neurology, Department of Medicine and Siriraj Neuroimmunology Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Mohammad A Sahraian
- MS Research Center, Neuroscience Institute, Teheran University of Medical Sciences, Iran
| | - Douglas K Sato
- Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Deanna Saylor
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; University Teaching Hospital, Lusaka, Zambia
| | - Fu-Dong Shi
- Tianjin Medical University General Hospital, Tianjin, China; National Clinical Research Center for Neurological Disorders, Beijing Tiantan Hospital, Beijing, China
| | - Aksel Siva
- Istanbul University Cerrahpasa, School of Medicine, Department of Neurology, Clinical Neuroimmunology Unit and MS Clinic, Istanbul, Türkiye
| | - Kevin Tan
- Department of Neurology, National Neuroscience Institute, Singapore; Duke-NUS Medical School, Singapore
| | | | - Mike P Wattjes
- Department of Neuroradiology, Charité Berlin, Corporate Member of Freie Universität zu Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Brian Weinshenker
- Department of Neurology, University of Virginia, Charlottesville, VA, USA
| | - Bassem Yamout
- Neurology Institute, Harley Street Medical Center, Abu Dhabi, United Arab Emirates
| | - Kazuo Fujihara
- Department of Multiple Sclerosis Therapeutics, Fukushima Medical University School of Medicine and Multiple Sclerosis and Neuromyelitis Optica Center, Southern TOHOKU Research Institute for Neuroscience, Koriyama, Japan.
| |
Collapse
|
84
|
Xin Y, Zhou Y. Association between cytomegalovirus infection and dyslipidemia in US: an NHANES analysis. Eur J Clin Microbiol Infect Dis 2024; 43:1883-1897. [PMID: 39066967 DOI: 10.1007/s10096-024-04905-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 07/18/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND Cytomegalovirus (CMV) infection is widely prevalent worldwide, which may have relationship with dyslipidemia. The aim of this study is to explore the association between CMV infection and dyslipidemia. METHODS The total observed population of this study included 14,163 participants aged 6-49 years from 1999 to 2004 National Health and Nutritional Examination Surveys (NHANES). Immunoglobulin G (IgG) levels and four lipid parameters (triglyceride, low density lipoprotein-cholesterol (LDL-C), total cholesterol, and high density lipoprotein-cholesterol (HDL-C)) were analyzed by performing multiple logistic regression and subgroup analysis. RESULTS The median values of triglycerides, LDL-C and total cholesterol levels in the CMV positive group were higher than those in CMV negative group while a lower median value of HDL-C existed in positive group. After controlling for potential confounders (sex, age, race, country of birth, education, poverty-to-income ratio(PIR)), a close association between CMV infection and low HDL-C was observed, which persisted in the men aged 30-49 and women aged 12-19, 30-49. CONCLUSIONS CMV infection is related to dyslipidemia, and this association is more significant in the serum HDL-C. Further cohort studies and experimental evidences can be conducted to test this association and then guide clinical practice.
Collapse
Affiliation(s)
- Yiyang Xin
- Department of Pathogen Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Yun Zhou
- Department of Pathogen Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China.
| |
Collapse
|
85
|
Pavia G, Licata F, Marascio N, Giancotti A, Tassone MT, Costa C, Scarlata GGM, Prestagiacomo LE, Gigliotti S, Trecarichi EM, Torti C, Bianco A, Quirino A, Matera G. Seroprevalence and age-related susceptibility of TORCH infections in childbearing age women: A 5-year cross-sectional retrospective study and a literature review. J Infect Public Health 2024; 17:102537. [PMID: 39255545 DOI: 10.1016/j.jiph.2024.102537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 08/23/2024] [Accepted: 08/28/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND Serodiagnosis of TORCH infections should be performed in pre-pregnancy and reproductive-age women to prevent vertical transmission. Herein, we conducted a 5-year cross-sectional retrospective study in childbearing age women to provide prevalence data. Also, stratifying the cohort into three age groups, we identified those most susceptible to acute TORCH infections. METHODS Between 2019 and 2023, serum samples from 2286 childbearing age women attending the "R. Dulbecco" University Hospital of Catanzaro were collected. Screening for TORCH pathogens, such as: Toxoplasma gondii (TOX), Cytomegalovirus (CMV), Rubella Virus (RUB), Parvovirus B19 (ParvoB19), Herpes Simplex Virus types 1 and 2 (HSV1, HSV2) and Treponema pallidum was carried out using serological tests. Chemiluminescent immunoassay was performed to detect TOX, CMV and ParvoB19 Immunoglobulin M (IgM) and Immunoglobulin G (IgG) antibodies, while Enzyme Linked Fluorescent Assay was performed to detect RUB IgM and IgG antibodies and CMV and TOX IgG Avidity. Enzyme Linked Immunosorbent Assay was performed to detect HSV1 IgG, HSV2 IgG, HSV1/2 IgM, T. pallidum total antibodies and RUB IgG Avidity. Binomial logistic regression models were developed to compare seroprevalence rates among different age groups. RESULTS The highest immunological protection was observed for RUB infection (87 %), probably associated with vaccination practice, followed by HSV1 and CMV (82 % and 63 %). The 16-25 year age group results as the most susceptible to acute infections as demonstrated by odds of CMV IgM positivity (primary infection) which decreased with age. CONCLUSIONS The TORCH serological screening program should be implemented in women before pregnancy to formulate strategies for serological screening of childbearing age women and guiding clinicians in making decisions.
Collapse
Affiliation(s)
- Grazia Pavia
- Unit of Clinical Microbiology, Department of Health Sciences, "Magna Græcia" University Hospital, 88100 Catanzaro, Italy
| | - Francesca Licata
- Department of Health Sciences, School of Medicine, "Magna Græcia" University Hospital, 88100 Catanzaro, Italy
| | - Nadia Marascio
- Unit of Clinical Microbiology, Department of Health Sciences, "Magna Græcia" University Hospital, 88100 Catanzaro, Italy.
| | - Aida Giancotti
- Unit of Clinical Microbiology, Department of Health Sciences, "Magna Græcia" University Hospital, 88100 Catanzaro, Italy
| | - Maria Teresa Tassone
- Infectious and Tropical Disease Unit, Department of Medical and Surgical Sciences, "Magna Graecia" University, 88100 Catanzaro, Italy
| | - Chiara Costa
- Infectious and Tropical Disease Unit, Department of Medical and Surgical Sciences, "Magna Graecia" University, 88100 Catanzaro, Italy
| | - Giuseppe Guido Maria Scarlata
- Unit of Clinical Microbiology, Department of Health Sciences, "Magna Græcia" University Hospital, 88100 Catanzaro, Italy
| | - Licia Elvira Prestagiacomo
- Unit of Clinical Microbiology, Department of Health Sciences, "Magna Græcia" University Hospital, 88100 Catanzaro, Italy
| | - Simona Gigliotti
- Unit of Clinical Microbiology, Department of Health Sciences, "Magna Græcia" University Hospital, 88100 Catanzaro, Italy
| | - Enrico Maria Trecarichi
- Infectious and Tropical Disease Unit, Department of Medical and Surgical Sciences, "Magna Graecia" University, 88100 Catanzaro, Italy
| | - Carlo Torti
- Infectious and Tropical Disease Unit, Department of Medical and Surgical Sciences, "Magna Graecia" University, 88100 Catanzaro, Italy
| | - Aida Bianco
- Department of Health Sciences, School of Medicine, "Magna Græcia" University Hospital, 88100 Catanzaro, Italy
| | - Angela Quirino
- Unit of Clinical Microbiology, Department of Health Sciences, "Magna Græcia" University Hospital, 88100 Catanzaro, Italy
| | - Giovanni Matera
- Unit of Clinical Microbiology, Department of Health Sciences, "Magna Græcia" University Hospital, 88100 Catanzaro, Italy
| |
Collapse
|
86
|
Gao F, Mora MC, Constantinides M, Coënon L, Multrier C, Vaillant L, Peyroux J, Zhang T, Villalba M. Feeder cell training shapes the phenotype and function of in vitro expanded natural killer cells. MedComm (Beijing) 2024; 5:e740. [PMID: 39314886 PMCID: PMC11417427 DOI: 10.1002/mco2.740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 08/25/2024] [Accepted: 08/26/2024] [Indexed: 09/25/2024] Open
Abstract
Natural killer (NK) cells are candidates for adoptive cell therapy, and the protocols for their activation and expansion profoundly influence their function and fate. The complexity of NK cell origin and feeder cell cues impacts the heterogeneity of expanded NK (eNK) cells. To explore this, we compared the phenotype and function of peripheral blood-derived NK (PB-NK) and umbilical cord blood-derived NK (UCB-NK) cells activated by common feeder cell lines, including K562, PLH, and 221.AEH. After first encounter, most PB-NK cells showed degranulation independently of cytokines production. Meanwhile, most UCB-NK cells did both. We observed that each feeder cell line uniquely influenced the activation, expansion, and ultimate fate of PB eNK and UCB eNK cells, determining whether they became cytokine producers or killer cells. In addition, they also affected the functional performance of NK cell subsets after expansion, that is, expanded conventional NK (ecNK) and expanded FcRγ- NK (eg-NK) cells. Hence, the regulation of eNK cell function largely depends on the NK cell source and the chosen expansion system. These results underscore the significance of selecting feeder cells for NK cell expansion from various sources, notably for customized adoptive cell therapies to yield cytokine-producing or cytotoxic eNK cells.
Collapse
Affiliation(s)
- Fei Gao
- IRMBUniversity of MontpellierINSERMCHR MontpellierMontpellierFrance
- Department of PathologySchool of Basic MedicineCentral South UniversityChangshaChina
| | | | | | - Loïs Coënon
- IRMBUniversity of MontpellierINSERMCHR MontpellierMontpellierFrance
| | | | - Loïc Vaillant
- IRMBUniversity of MontpellierINSERMCHR MontpellierMontpellierFrance
| | - Julien Peyroux
- IRMBUniversity of MontpellierINSERMCHR MontpellierMontpellierFrance
| | - Tianxiang Zhang
- Department of ImmunobiologyYale University School of MedicineNew HavenConnecticutUSA
| | - Martin Villalba
- IRMBUniversity of MontpellierINSERMCHR MontpellierMontpellierFrance
- Institut du Cancer Avignon‐Provence Sainte CatherineAvignonFrance
- IRMBUniv MontpellierINSERMCHU MontpellierCNRSMontpellierFrance
| |
Collapse
|
87
|
Puopolo KM. Postnatally acquired cytomegalovirus infection among preterm infants. Curr Opin Infect Dis 2024; 37:425-430. [PMID: 39105629 DOI: 10.1097/qco.0000000000001047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/07/2024]
Abstract
PURPOSE OF REVIEW Although there are multiple benefits of mother's own milk feeding for very-low birth weight, low gestation infants, those born to cytomegalovirus (CMV)-seropositive mothers are at risk for acquiring postnatal CMV infection. This review will describe the risk and consequences of postnatal CMV infection among very preterm infants. RECENT FINDINGS Postnatal CMV may manifest as clinically silent infection or as mild to severe and occasionally fatal disease. The risk of disease is balanced by the health benefits of human milk feeding to preterm infants. Postnatal CMV infection has been associated with increased risks of multiple preterm morbidities such as bronchopulmonary dysplasia, necrotizing enterocolitis and neurodevelopmental impairment, but current evidence is limited by the selection bias inherent to reporting in case series and retrospective cohort studies. SUMMARY Knowledge gaps exist regarding the risk-benefit balance of pasteurization to inactivate CMV in fresh breast milk, as well as the optimal dosing, duration and efficacy of treating infected infants with antiviral medications. Multicenter, prospective studies are urgently needed to accurately determine the true burden that postnatal CMV infection presents to very preterm infants. Such studies will inform the need for preventive strategies and treatment guidance.
Collapse
Affiliation(s)
- Karen M Puopolo
- Division of Neonatology and Clinical Futures, Children's Hospital of Philadelphia
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine
- Section on Newborn Medicine, Pennsylvania Hospital, Philadelphia, Pennsylvania, USA
| |
Collapse
|
88
|
Sayyed A, Wilson L, Stavi V, Chen S, Chen C, Mattsson J, Lipton JH, Kim DD, Viswabandya A, Kumar R, Lam W, Law AD, Gerbitz A, Pasic I, Novitzky-Basso I, Mazzulli T, Michelis FV. Impact of cytomegalovirus (CMV) seroconversion pre-allogeneic hematopoietic cell transplantation on posttransplant outcomes. Eur J Haematol 2024; 113:441-453. [PMID: 38880946 DOI: 10.1111/ejh.14251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 06/18/2024]
Abstract
Cytomegalovirus (CMV) reactivation post-allogeneic hematopoietic cell transplantation (post-alloHCT) increases morbidity and mortality. We sought to determine the frequency of CMV seroconversion in patients pre-alloHCT and to investigate the impact on posttransplant outcomes. We retrospectively investigated 752 adult patients who underwent alloHCT at our center from January 2015 to February 2020 before the adoption of letermovir prophylaxis. CMV serology was assessed at consult and pretransplant. The cohort was divided into four groups based on pretransplant CMV seroconversion: negative to positive (Group 1), positive to negative (Group 2), consistently negative (Group 3), and consistently positive (Group 4). Eighty-nine patients (12%) had seroconverted from negative to positive, 17 (2%) from positive to negative, 151 (20%) were consistently seronegative, and 495 (66%) were consistently seropositive pretransplant. For the four CMV serostatus groups, cumulative incidence of CMV reactivation at 6 months posttransplant was 4.5%, 47.1%, 6.6%, and 76.6% for Groups 1, 2, 3, and 4, respectively (p < .0001). No differences between groups were seen regarding Grade III-IV acute graft-versus-host disease (GVHD) (p = .91), moderate/severe chronic GVHD (p = .41), or graft failure (p = .28). On multivariable analysis, there was no impact of CMV serostatus group on overall survival (p = .67), cumulative incidence of relapse (p = .83) or non-relapse mortality. alloHCT patients who demonstrate CMV seroconversion pretransplant from negative to positive have a very low risk of CMV reactivation posttransplant. The observed seroconversion may be due to passive CMV immunity acquired through blood products. Quantitative CMV immunoglobulin G/immunoglobulin M pretransplant may help differentiate between true seroconversion and passively transmitted CMV immunoglobulin.
Collapse
Affiliation(s)
- Ayman Sayyed
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Leeann Wilson
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Vered Stavi
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Shiyi Chen
- Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Carol Chen
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Jonas Mattsson
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Jeffrey H Lipton
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Dennis D Kim
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Auro Viswabandya
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Rajat Kumar
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Wilson Lam
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Arjun D Law
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Armin Gerbitz
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Ivan Pasic
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Igor Novitzky-Basso
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Tony Mazzulli
- Department of Microbiology, Sinai Health System/University Health Network, Toronto, Ontario, Canada
| | - Fotios V Michelis
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
89
|
Piña A, Elko EA, Caballero R, Metrailer M, Mulrow M, Quan D, Nordstrom L, Altin JA, Ladner JT. Mapping disparities in viral infection rates using highly multiplexed serology. mSphere 2024; 9:e0012724. [PMID: 39162531 PMCID: PMC11423740 DOI: 10.1128/msphere.00127-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 07/21/2024] [Indexed: 08/21/2024] Open
Abstract
Despite advancements in medical interventions, the disease burden caused by viral pathogens remains large and highly diverse. This burden includes the wide range of signs and symptoms associated with active viral replication as well as a variety of clinical sequelae of infection. Moreover, there is growing evidence supporting the existence of sex- and ethnicity-based health disparities linked to viral infections and their associated diseases. Despite several well-documented disparities in viral infection rates, our current understanding of virus-associated health disparities remains incomplete. This knowledge gap can be attributed, in part, to limitations of the most commonly used viral detection methodologies, which lack the breadth needed to characterize exposures across the entire virome. Additionally, virus-related health disparities are dynamic and often differ considerably through space and time. In this study, we utilize PepSeq, an approach for highly multiplexed serology, to broadly assess an individual's history of viral exposures, and we demonstrate the effectiveness of this approach for detecting infection disparities through a pilot study of 400 adults aged 30-60 in Phoenix, AZ. Using a human virome PepSeq library, we observed expected seroprevalence rates for several common viruses and detected both expected and previously undocumented differences in inferred rates of infection between our male/female and Hispanic/non-Hispanic White individuals. IMPORTANCE Our understanding of population-level virus infection rates and associated health disparities is incomplete. In part, this is because of the high diversity of human-infecting viruses and the limited breadth and sensitivity of traditional approaches for detecting infection events. Here, we demonstrate the potential for modern, highly multiplexed antibody detection methods to greatly increase our understanding of disparities in rates of infection across subpopulations (e.g., different sexes or ethnic groups). The use of antibodies as biomarkers allows us to detect evidence of past infections over an extended period, and our approach for highly multiplexed serology (PepSeq) allows us to measure antibody responses against hundreds of viruses in an efficient and cost-effective manner.
Collapse
Affiliation(s)
- Alejandra Piña
- The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, Arizona, USA
| | - Evan A Elko
- The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, Arizona, USA
| | | | - Morgan Metrailer
- The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, Arizona, USA
| | | | - Dan Quan
- Valleywise Health, Phoenix, Arizona, USA
- University of Arizona, College of Medicine, Phoenix, Arizona, USA
- Creighton University, School of Medicine, Phoenix, Arizona, USA
| | | | - John A Altin
- The Translational Genomics Research Institute (TGen), Flagstaff, Arizona, USA
| | - Jason T Ladner
- The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, Arizona, USA
| |
Collapse
|
90
|
Freeman MR, Dooley AL, Beucler MJ, Sanders W, Moorman NJ, O'Connor CM, Miller WE. The Human Cytomegalovirus vGPCR UL33 is Essential for Efficient Lytic Replication in Epithelial Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.18.609710. [PMID: 39345593 PMCID: PMC11429895 DOI: 10.1101/2024.09.18.609710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Human cytomegalovirus (HCMV) is a β-herpesvirus which is ubiquitous in the human population. HCMV has the largest genome of all known human herpesviruses, and thus encodes a large array of proteins that affect pathogenesis in different cell types. Given the large genome and the ability of HCMV to replicate in a range of cells, investigators have begun to identify viral proteins required for cell type-specific replication. There are four proteins encoded in the HCMV genome that are homologous to human G protein-coupled receptors (GPCRs); these viral-encoded GPCRs (vGPCRs) are UL33, UL78, US27, and US28. In the current study, we find that deletion of all four vGPCR genes from a clinical isolate of HCMV severely attenuates lytic replication in both primary human salivary gland epithelial cells, as well as ARPE-19 retinal epithelial cells as evidenced by significant decreases in immediate early gene expression and virus production. Deletion of UL33 from the HCMV genome also results in a failure to efficiently replicate in epithelial cells, and this defect is manifested by decreased levels of immediate early, early, and late gene expression, as well as reduced viral production. We find that similar to US28, UL33 constitutively activates Gαq-dependent PLC-β signaling to high levels in these epithelial cells. We also find that UL33 transcription is more complicated than originally believed, and there is the potential for the virus to utilize various 5' UTRs to create novel UL33 proteins that are all capable of constitutive Gαq signaling. Taken together, these studies suggest that UL33 driven signaling is important for lytic HCMV replication in cells of epithelial origin.
Collapse
|
91
|
Karner D, Kvestak D, Kucan Brlic P, Cokaric Brdovcak M, Lisnic B, Brizic I, Juranic Lisnic V, Golemac M, Tomac J, Krmpotic A, Karkeni E, Libri V, Mella S, Legname G, Altmeppen HC, Hasan M, Jonjic S, Lenac Rovis T. Prion protein alters viral control and enhances pathology after perinatal cytomegalovirus infection. Nat Commun 2024; 15:7754. [PMID: 39237588 PMCID: PMC11377837 DOI: 10.1038/s41467-024-51931-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 08/20/2024] [Indexed: 09/07/2024] Open
Abstract
Cytomegalovirus (CMV) infection poses risks to newborns, necessitating effective therapies. Given that the damage includes both viral infection of brain cells and immune system-related damage, here we investigate the involvement of cellular prion protein (PrP), which plays vital roles in neuroprotection and immune regulation. Using a murine model, we show the role of PrP in tempering neonatal T cell immunity during CMV infection. PrP-null mice exhibit enhanced viral control through elevated virus-specific CD8 T cell responses, leading to reduced viral titers and pathology. We further unravel the molecular mechanisms by showing CMV-induced upregulation followed by release of PrP via the metalloproteinase ADAM10, impairing CD8 T cell response specifically in neonates. Additionally, we confirm PrP downregulation in human CMV (HCMV)-infected fibroblasts, underscoring the broader relevance of our observations beyond the murine model. Furthermore, our study highlights how PrP, under the stress of viral pathogenesis, reveals its impact on neonatal immune modulation.
Collapse
Affiliation(s)
- Dubravka Karner
- Center for Proteomics; Faculty of Medicine; University of Rijeka, Rijeka, Croatia
| | - Daria Kvestak
- Center for Proteomics; Faculty of Medicine; University of Rijeka, Rijeka, Croatia
| | - Paola Kucan Brlic
- Center for Proteomics; Faculty of Medicine; University of Rijeka, Rijeka, Croatia
| | | | - Berislav Lisnic
- Center for Proteomics; Faculty of Medicine; University of Rijeka, Rijeka, Croatia
| | - Ilija Brizic
- Center for Proteomics; Faculty of Medicine; University of Rijeka, Rijeka, Croatia
| | - Vanda Juranic Lisnic
- Center for Proteomics; Faculty of Medicine; University of Rijeka, Rijeka, Croatia
| | - Mijo Golemac
- Department of Histology and Embryology; Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Jelena Tomac
- Department of Histology and Embryology; Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Astrid Krmpotic
- Department of Histology and Embryology; Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Esma Karkeni
- Cytometry and Biomarkers Unit of Technology and Service (CB TechS); Institut Pasteur, Université Paris Cité, Paris, France
| | - Valentina Libri
- Cytometry and Biomarkers Unit of Technology and Service (CB TechS); Institut Pasteur, Université Paris Cité, Paris, France
| | - Sebastien Mella
- Cytometry and Biomarkers Unit of Technology and Service (CB TechS); Institut Pasteur, Université Paris Cité, Paris, France
| | - Giuseppe Legname
- Department of Neuroscience, Prion Biology Laboratory, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Trieste, Italy
| | - Hermann C Altmeppen
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Milena Hasan
- Cytometry and Biomarkers Unit of Technology and Service (CB TechS); Institut Pasteur, Université Paris Cité, Paris, France
| | - Stipan Jonjic
- Center for Proteomics; Faculty of Medicine; University of Rijeka, Rijeka, Croatia
| | - Tihana Lenac Rovis
- Center for Proteomics; Faculty of Medicine; University of Rijeka, Rijeka, Croatia.
| |
Collapse
|
92
|
Mendes I, Vara-Luiz F, Carralas Antunes S, Nunes G. Recurrent gastrointestinal bleeding caused by cytomegalovirus duodenitis. REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2024; 118. [PMID: 39235203 DOI: 10.17235/reed.2024.10678/2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/06/2024]
Abstract
A 78-year-old male hospitalized due to acute pneumonia presented with hematemesis, melena and hypotension. Past medical history was relevant for prostate cancer with bone and lung metastasis under systemic chemotherapy. Upper gastrointestinal endoscopy revealed a 50mm serpiginous ulcer in the second portion of the duodenum with a visible vessel (Forrest IIa). Hemostatic therapy with adrenaline, polidocanol and one clip placement was performed. Biopsies of the ulcer were also obtained. During hospitalization the patient developed recurrent gastrointestinal bleeding requiring several red blood cell transfusions and endoscopic reintervention. Further histopathologic evaluation revealed chronic duodenitis caused by cytomegalovirus (CMV) infection. Considering the poor performance status and successful hemostasis with ulcer healing, antiviral treatment was not initiated. The patient died a few weeks later due to neoplastic disease progression.
Collapse
Affiliation(s)
- Ivo Mendes
- Gastroenterology , Hospital Garcia de Orta, Portugal
| | | | | | | |
Collapse
|
93
|
Vietzen H, Simonitsch C, Friedel B, Berger SM, Kühner LM, Furlano PL, Florian DM, Görzer I, Koblischke M, Aberle JH, Puchhammer-Stöckl E. Torque teno viruses exhaust and imprint the human immune system via the HLA-E/NKG2A axis. Front Immunol 2024; 15:1447980. [PMID: 39295866 PMCID: PMC11408220 DOI: 10.3389/fimmu.2024.1447980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/09/2024] [Indexed: 09/21/2024] Open
Abstract
The ubiquitous Torque teno virus (TTV) establishes a chronically persistent infection in the human host. TTV has not been associated with any apparent disease, but, as part of the human virome, it may confer a regulatory imprint on the human immune system with as yet unclear consequences. However, so far, only few studies have characterized the TTV-specific immune responses or the overall immunological imprints by TTV. Here, we reveal that TTV infection leads to a highly exhausted TTV-specific CD8+ T-cell response, hallmarked by decreased IFN-γ production and the expression of the inhibitory NKG2A-receptor. On a functional level, we identified a panel of highly polymorphic TTV-encoded peptides that lead to an expansion of regulatory NKG2A+ natural killer, NKG2A+CD4+, and NKG2A+CD8+ T cells via the stabilization of the non-classical HLA-E molecule. Our results thus demonstrate that TTV leads to a distinct imprint on the human immune system that may further regulate overall human immune responses in infectious, autoimmune, and malignant diseases.
Collapse
Affiliation(s)
- Hannes Vietzen
- Center for Virology, Medical University Vienna, Vienna, Austria
| | - Cara Simonitsch
- Center for Virology, Medical University Vienna, Vienna, Austria
| | | | - Sarah M Berger
- Center for Virology, Medical University Vienna, Vienna, Austria
| | - Laura M Kühner
- Center for Virology, Medical University Vienna, Vienna, Austria
| | | | - David M Florian
- Center for Virology, Medical University Vienna, Vienna, Austria
| | - Irene Görzer
- Center for Virology, Medical University Vienna, Vienna, Austria
| | | | - Judith H Aberle
- Center for Virology, Medical University Vienna, Vienna, Austria
| | | |
Collapse
|
94
|
Pontes KFM, Araujo E. Cytomegalovirus and pregnancy: current evidence for clinical practice. REVISTA DA ASSOCIACAO MEDICA BRASILEIRA (1992) 2024; 70:e20240509. [PMID: 39230148 PMCID: PMC11370743 DOI: 10.1590/1806-9282.20240509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 04/19/2024] [Indexed: 09/05/2024]
Affiliation(s)
- Karina Felippe Monezi Pontes
- Universidade Federal de São Paulo, Paulista School of Medicine, Department of Obstetrics – São Paulo (SP), Brazil
- Ipiranga Hospital, Service of Gynecology and Obstetrics – São Paulo (SP), Brazil
| | - Edward Araujo
- Universidade Federal de São Paulo, Paulista School of Medicine, Department of Obstetrics – São Paulo (SP), Brazil
- Universidade Municipal de São Caetano do Sul, Discipline of Woman Health – São Caetano do Sul (SP), Brazil
| |
Collapse
|
95
|
Ekman I, Schroderus AM, Vuorinen T, Knip M, Veijola R, Toppari J, Ilonen J, Lempainen J, Kinnunen T. The effect of early life cytomegalovirus infection on the immune profile of children. Clin Immunol 2024; 266:110330. [PMID: 39067678 DOI: 10.1016/j.clim.2024.110330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/05/2024] [Accepted: 07/20/2024] [Indexed: 07/30/2024]
Abstract
Cytomegalovirus (CMV) infection has a life-long impact on the immune system, particularly on memory T cells. However, the effect of early life CMV infection on the phenotype and functionality of T cells in infants and especially longitudinal changes occurring during childhood have not been explored in detail. The phenotype and functionality of peripheral blood CD8+ and CD4+ T cells from children infected with CMV in early life (< 6 months of age) was analyzed using high-dimensional flow cytometry. Samples from CMV IgG-seropositive (CMV+) children were collected at 6 months and 6 years of age and compared to samples from CMV-seronegative (CMV-) children. Early life CMV infection caused multiple alterations within T cells. These include downregulation of CD28 expression and upregulation of CD57 expression within both CD27+ early and CD27- late effector memory CD8+ and CD4+ T-cells at 6 months of age. Of these changes, only alterations within the highly differentiated late effector memory compartment persisted at the age of 6 years. Early life CMV-infection has a distinct impact on developing CD8+ and CD4+ memory T cell compartments. It appears to induce both temporary as well as longer-lasting alterations, which may affect the functionality of the immune system throughout life.
Collapse
Affiliation(s)
- Ilse Ekman
- Department of Clinical Microbiology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Anna-Mari Schroderus
- Department of Clinical Microbiology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Tytti Vuorinen
- Institute of Biomedicine, University of Turku, Turku, Finland; Department of Clinical Microbiology, Turku University Hospital, Turku, Finland
| | - Mikael Knip
- Tampere Center for Child Health Research, Tampere University Hospital, Tampere, Finland; Research Unit of Clinical Medicine, Department of Pediatrics, Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Riitta Veijola
- PEDEGO Research Unit, Department of Pediatrics, Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Jorma Toppari
- Department of Pediatrics, University of Turku and Turku University Hospital, Turku, Finland; Research Centre for Integrative Physiology and Pharmacology, and Centre for Population Health Research, InFLAMES Research Flagship, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Jorma Ilonen
- Immunogenetics Laboratory, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Johanna Lempainen
- Department of Clinical Microbiology, Turku University Hospital, Turku, Finland; Department of Pediatrics, University of Turku and Turku University Hospital, Turku, Finland; Immunogenetics Laboratory, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Tuure Kinnunen
- Department of Clinical Microbiology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland; ISLAB Laboratory Centre, Kuopio, Finland.
| |
Collapse
|
96
|
Sundi A, Roopa N, Gupta P, Irfan S, Kar TK. Seroprevalent study of cytomegalovirus infection in the regions of Jharkhand. J Family Med Prim Care 2024; 13:4066-4070. [PMID: 39464971 PMCID: PMC11504779 DOI: 10.4103/jfmpc.jfmpc_259_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/26/2024] [Accepted: 06/21/2024] [Indexed: 10/29/2024] Open
Abstract
Background The Human Cytomegalovirus (HCMV) is a type of beta herpesvirus widespread in all human populations. It is estimated that up to 80-100% of adults worldwide and most infections are harmless and can cause severe health complications in infants, like hearing loss and developmental issues. Still, immunocompromised individuals can experience serious complications from the virus. Unfortunately, there is limited information on the prevalence of this virus in our country, and no studies have been reported on the rate of CMV transmission yet. Objectives This study aims to evaluate the levels of IgM antibodies against Cytomegalovirus (CMV) in East Singhbhum, West Singhbhum, and Seraikela Kharsawan using an ELISA test. Methods An indirect ELISA test was performed to detect anti-CMV IgM and the period of study was from January'2021 to June'2023. Results The examination tested 55 people for the TORCH profile of CMV parameters from regions of East Singhbhum, West Singhbhum, and Seraikela Kharsawan. Here, 17 people (30.09%) were IgM positive by ELISA. Conclusions The serological data confirms that CMV is not being monitored and recognized in the general population, which limits our study between CMV infection, disease, and clinically diagnosed outcomes. This understanding is crucial for the healthcare and policy sectors. Thus, we recommend implementing a surveillance and mindfulness program for at least one-fourth of the population in Jharkhand and continuing to explore and develop effective vaccines to control CMV infections.
Collapse
Affiliation(s)
- Anita Sundi
- Department of Microbiology, Virus Research Diagnostic Laboratory, Mahatma Gandhi Memorial Medical College, Jamshedpur, Jharkhand, India
| | - Nilamber Roopa
- Department of Microbiology, Virus Research Diagnostic Laboratory, Mahatma Gandhi Memorial Medical College, Jamshedpur, Jharkhand, India
| | - Piyalee Gupta
- Department of Microbiology, Virus Research Diagnostic Laboratory, Mahatma Gandhi Memorial Medical College, Jamshedpur, Jharkhand, India
| | - Sana Irfan
- Department of Microbiology, Virus Research Diagnostic Laboratory, Mahatma Gandhi Memorial Medical College, Jamshedpur, Jharkhand, India
| | - Tarun Kumar Kar
- Department of Microbiology, Virus Research Diagnostic Laboratory, Mahatma Gandhi Memorial Medical College, Jamshedpur, Jharkhand, India
| |
Collapse
|
97
|
Ren X, Wang K, Chang Z, Liu M, Cheng F, Min B, Wei S. Serological Screening of TORCH Pathogen Infections in Infertile Women of Childbearing Age in Northwest China. Reprod Sci 2024; 31:2877-2884. [PMID: 38630173 DOI: 10.1007/s43032-024-01551-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 04/02/2024] [Indexed: 09/14/2024]
Abstract
Serological screening for TORCH(Toxoplasma gondii [TOX], Rubella virus [RV], Cytomegalovirus [CMV], and Herpes simplex virus [HSV]) infections is an effective method for preventing congenital infections caused by TORCH pathogens.In this study, we retrospectively analyzed the characteristics of TORCH infections in 17,807 infertile women of childbearing age in northwest China.We conducted serological detection of TORCH-pathogen-specific IgM and IgG antibodies. The seroprevalence of TORCH infections was statistically analyzed by applying χ2 and Fisher exact-probability tests to evaluate the differences among ages and across quarters of the year. The overall IgM/IgG seroprevalences of TOX, RV, CMV, HSV-1, and HSV-2 were 0.46/3.4%, 0.77/84.93%, 0.68/97.54%, 1.2/82.83%, and 0.62/10.04%, respectively. The positive rates for RV-IgM in women ≥ 40 years old were significantly higher than those for women 25-39 (P < 0.05) years of age. The seroprevalence of HSV1-IgM was higher in the third and fourth quarters of the year (seasons) (P < 0.001), and the seroprevalence of CMV-IgG was statistically significant between differences quarters (P = 0.017), and the seroprevalence of CMV-IgG in the first quarter was lower than that in the third and fourth quarters (Bonferroni correction, P = 0.009 > 0.0083), suggesting no statistically significant difference between the latter two groups. This study showed that in northwestern China the risk of acquiring primary infection by a TORCH pathogen among infertile women of childbearing age were still high, especially Toxoplasma gondii and Herpesvirus type 2 infection. Therefore, effective prevention strategies that include serological screening for TORCH should be implemented.
Collapse
Affiliation(s)
- Xiaoyan Ren
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, Tang Du Hospital, The Air Force Military Medical University, Xi'an, China
| | - Kaili Wang
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, Tang Du Hospital, The Air Force Military Medical University, Xi'an, China
| | - Zhenhua Chang
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, Tang Du Hospital, The Air Force Military Medical University, Xi'an, China
| | - Mengxin Liu
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, Tang Du Hospital, The Air Force Military Medical University, Xi'an, China
| | - Fang Cheng
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, Tang Du Hospital, The Air Force Military Medical University, Xi'an, China
| | - Baohua Min
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, Tang Du Hospital, The Air Force Military Medical University, Xi'an, China
| | - Sanhua Wei
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, Tang Du Hospital, The Air Force Military Medical University, Xi'an, China.
| |
Collapse
|
98
|
Ortigas-Vasquez A, Szpara M. Embracing Complexity: What Novel Sequencing Methods Are Teaching Us About Herpesvirus Genomic Diversity. Annu Rev Virol 2024; 11:67-87. [PMID: 38848592 DOI: 10.1146/annurev-virology-100422-010336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2024]
Abstract
The arrival of novel sequencing technologies throughout the past two decades has led to a paradigm shift in our understanding of herpesvirus genomic diversity. Previously, herpesviruses were seen as a family of DNA viruses with low genomic diversity. However, a growing body of evidence now suggests that herpesviruses exist as dynamic populations that possess standing variation and evolve at much faster rates than previously assumed. In this review, we explore how strategies such as deep sequencing, long-read sequencing, and haplotype reconstruction are allowing scientists to dissect the genomic composition of herpesvirus populations. We also discuss the challenges that need to be addressed before a detailed picture of herpesvirus diversity can emerge.
Collapse
Affiliation(s)
- Alejandro Ortigas-Vasquez
- Departments of Biology and of Biochemistry and Molecular Biology; Center for Infectious Disease Dynamics; and Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania, USA;
| | - Moriah Szpara
- Departments of Biology and of Biochemistry and Molecular Biology; Center for Infectious Disease Dynamics; and Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania, USA;
| |
Collapse
|
99
|
Zong Y, Kamoi K, Miyagaki M, Zhang J, Yang M, Zou Y, Ohno-Matsui K. Applications of Biological Therapy for Latent Infections: Benefits and Risks. Int J Mol Sci 2024; 25:9184. [PMID: 39273134 PMCID: PMC11394918 DOI: 10.3390/ijms25179184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
Biological therapies have revolutionized medical treatment by targeting the key mediators or receptors involved in inflammatory responses, thereby effectively suppressing inflammation and achieving beneficial outcomes. They are more advanced than conventional therapies using corticosteroids and immunosuppressants, offering effective solutions for autoimmune diseases, cancer, transplant rejection, and various infectious diseases, including coronavirus disease 2019. Although they exert low immunosuppressive effects, biological therapies can reactivate specific biological targets associated with infections. This review summarizes the currently available biological therapies and discusses their immunosuppressive mechanisms and clinical applications, highlighting the variations in the types and frequencies of infection recurrence induced by different biological agents. Additionally, this review describes the risk factors associated with various biological agents, thus aiding clinicians in selecting the most appropriate biological therapy.
Collapse
Affiliation(s)
- Yuan Zong
- Department of Ophthalmology & Visual Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Koju Kamoi
- Department of Ophthalmology & Visual Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Miki Miyagaki
- Department of Ophthalmology & Visual Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Jing Zhang
- Department of Ophthalmology & Visual Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Mingming Yang
- Department of Ophthalmology & Visual Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Yaru Zou
- Department of Ophthalmology & Visual Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Kyoko Ohno-Matsui
- Department of Ophthalmology & Visual Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| |
Collapse
|
100
|
Litvin U, Wang ECY, Stanton RJ, Fielding CA, Hughes J. Evolution of the Cytomegalovirus RL11 gene family in Old World monkeys and Great Apes. Virus Evol 2024; 10:veae066. [PMID: 39315401 PMCID: PMC11416908 DOI: 10.1093/ve/veae066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/15/2024] [Accepted: 08/22/2024] [Indexed: 09/25/2024] Open
Abstract
Cytomegalovirus (CMV) is a genus of herpesviruses, members of which share a long history of coevolution with their primate hosts including New World monkeys, Old World monkeys (OWMs), and Great Apes (GAs). These viruses are ubiquitous within their host populations and establish lifelong infection in most individuals. Although asymptomatic in healthy individuals, infection poses a significant risk to individuals with a weakened or underdeveloped immune system. The genome of human CMV is the largest among human-infecting viruses and comprises at least 15 separate gene families, which may have arisen by gene duplication. Within human CMV, the RL11 gene family is the largest. RL11 genes are nonessential in vitro but have immune evasion roles that are likely critical to persistence in vivo. These genes demonstrate an extreme level of inter-species and intra-strain sequence diversity, which makes it challenging to deduce the evolutionary relationships within this gene family. Understanding the evolutionary relationships of these genes, especially accurate ortholog identification, is essential for reconstructing ancestral genomes, deciphering gene repertoire and order, and enabling reliable functional analyses across the CMV species, thereby offering insights into evolutionary processes, genetic diversity, and the functional significance of genes. In this work, we combined in silico genome screening with sequence-based and structure-guided phylogenetic analysis to reconstruct the evolutionary history of the RL11 gene family. We confirmed that RL11 genes are unique to OWM and GA CMVs, showing that this gene family was formed by multiple early duplication events and later lineage-specific losses. We identified four main clades of RL11 genes and showed that their expansions were mainly lineage specific and happened independently in CMVs of GAs, African OWMs, and Asian OWMs. We also identified groups of orthologous genes across the CMV tree, showing that some human CMV-specific RL11 genes emerged before the divergence of human and chimpanzee CMVs but were subsequently lost in the latter. The extensive and dynamic species-specific evolution of this gene family suggests that their functions target elements of host immunity that have similarly coevolved during speciation.
Collapse
Affiliation(s)
- Ulad Litvin
- MRC-University of Glasgow Centre for Virus Research, Sir Michael Stoker Building, 464 Bearsden Road, Glasgow G61 1QH, United Kingdom
| | - Eddie C Y Wang
- Division of Infection and Immunity, Cardiff University School of Medicine, UHW Main Building, Heath Park, Cardiff CF14 4XN, United Kingdom
| | - Richard J Stanton
- Division of Infection and Immunity, Cardiff University School of Medicine, UHW Main Building, Heath Park, Cardiff CF14 4XN, United Kingdom
| | - Ceri A Fielding
- Division of Infection and Immunity, Cardiff University School of Medicine, UHW Main Building, Heath Park, Cardiff CF14 4XN, United Kingdom
| | - Joseph Hughes
- MRC-University of Glasgow Centre for Virus Research, Sir Michael Stoker Building, 464 Bearsden Road, Glasgow G61 1QH, United Kingdom
| |
Collapse
|